51
|
Huang H, Li N, Wei X, Li Q, Guo J, Yang G, Yang H, Cai L, Liu Y, Wu C. Biomimetic "Gemini nanoimmunoregulators" orchestrated for boosted photoimmunotherapy by spatiotemporally modulating PD-L1 and tumor-associated macrophages. Acta Pharm Sin B 2024; 14:1345-1361. [PMID: 38486995 PMCID: PMC10935025 DOI: 10.1016/j.apsb.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 03/17/2024] Open
Abstract
A novel strategy of not only stimulating the immune cycle but also modulating the immunosuppressive tumor microenvironment is of vital importance to efficient cancer immunotherapy. Here, a new type of spatiotemporal biomimetic "Gemini nanoimmunoregulators" was engineered to activate robust systemic photoimmunotherapy by integrating the triple-punch of amplified immunogenic cell death (ICD), tumor-associated macrophages (TAMs) phenotype reprogramming and programmed cell death ligand 1 (PD-L1) degradation. The "Gemini nanoimmunoregulators" PM@RM-T7 and PR@RM-M2 were constructed by taking the biocompatible mesoporous polydopamine (mPDA) as nanovectors to deliver metformin (Met) and toll-like receptor 7/8 agonist resiquimod (R848) to cancer cells and TAMs by specific biorecognition via wrapping of red blood cell membrane (RM) inlaid with T7 or M2 peptides. mPDA/Met@RM-T7 (abbreviated as PM@RM-T7) was constructed to elicit an amplified in situ ICD effect through the targeted PTT and effectively stimulated the anticancer immunity. Meanwhile, PD-L1 on the remaining cancer cells was degraded by the burst metformin to prevent immune evasion. Subsequently, mPDA/R848@RM-M2 (abbreviated as PR@RM-M2) specifically recognized TAMs and reset the phenotype from M2 to M1 state, thus disrupting the immunosuppressive microenvironment and further boosting the function of cytotoxic T lymphocytes. This pair of sister nanoimmunoregulators cooperatively orchestrated the comprehensive anticancer activity, which remarkably inhibited the growth of primary and distant 4T1 tumors and prevented malignant metastasis. This study highlights the spatiotemporal cooperative modalities using multiple nanomedicines and provides a new paradigm for efficient cancer immunotherapy against metastatic-prone tumors.
Collapse
Affiliation(s)
- Honglin Huang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ningxi Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xiaodan Wei
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Qingzhi Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Junhan Guo
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Geng Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hong Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Lulu Cai
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yiyao Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Chunhui Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
52
|
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
|
53
|
Wang S, Wang J, Chen Z, Luo J, Guo W, Sun L, Lin L. Targeting M2-like tumor-associated macrophages is a potential therapeutic approach to overcome antitumor drug resistance. NPJ Precis Oncol 2024; 8:31. [PMID: 38341519 DOI: 10.1038/s41698-024-00522-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Tumor drug resistance emerges from the interaction of two critical factors: tumor cellular heterogeneity and the immunosuppressive nature of the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) constitute essential components of the TME. M2-like TAMs are essential in facilitating tumor metastasis as well as augmenting the drug resistance of tumors. This review encapsulates the mechanisms that M2-like TAMs use to promote tumor drug resistance. We also describe the emerging therapeutic strategies that are currently targeting M2-like TAMs in combination with other antitumor drugs, with some still undergoing clinical trial evaluation. Furthermore, we summarize and analyze various existing approaches for developing novel drugs that target M2-like TAMs to overcome tumor resistance, highlighting how targeting M2-like TAMs can effectively stop tumor growth, metastasis, and overcome tumor drug resistance.
Collapse
Affiliation(s)
- Shujing Wang
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingrui Wang
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiqiang Chen
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiamin Luo
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Guo
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Sun
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhu Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
54
|
Guo X, Wang P, Li Y, Chang Y, Wang X. Microbiomes in pancreatic cancer can be an accomplice or a weapon. Crit Rev Oncol Hematol 2024; 194:104262. [PMID: 38199428 DOI: 10.1016/j.critrevonc.2024.104262] [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: 08/31/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024] Open
Abstract
Recently, several investigations have linked the microbiome to pancreatic cancer progression. It is critical to reveal the role of different microbiomes in the occurrence, development, and treatment of pancreatic cancer. The current review summarizes the various microbiota types in pancreatic cancer while updating and supplementing the mechanisms of the representative gut, pancreatic, and oral microbiota, and their metabolites during its pathogenesis and therapeutic intervention. Several novel strategies have been introduced based on the tumor-associated microbiome to optimize the early diagnosis and prognosis of pancreatic cancer. The pros and cons involving different microbiomes in treating pancreatic cancer are discussed. The microbiome-related clinical trials for pancreatic cancer theranostics are outlined. This convergence of cutting-edge knowledge will provide feasible ideas for developing innovative therapies against pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoyu Guo
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Pan Wang
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yuan Li
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yawei Chang
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xiaobing Wang
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
55
|
Zhao X, Zheng C, Wang Y, Hao J, Liu Y. GSH/pH dual responsive chitosan nanoparticles for reprogramming M2 macrophages and overcoming cancer chemoresistance. Biomater Sci 2024; 12:790-797. [PMID: 38179727 DOI: 10.1039/d3bm01741a] [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: 01/06/2024]
Abstract
The combination of two or more drugs with different mechanisms of action is a promising strategy for circumventing multidrug resistance (MDR). However, the antitumor effect of nanosystems is usually limited due to the simultaneous release of different payloads at a single location rather than at their respective sites of action. Herein, we report a GSH and pH dual responsive nanoplatform encapsulated with doxorubicin (DOX) and resiquimod (R848) (GPNP) for combinatorial chemotherapy against cancer cells with drug resistance. GPNP possesses a core-shell structure wherein the polymer shell detaches in the acidic and sialic acid (SA)-rich environment. This leads to the release of R848 into the tumor microenvironment (TME), thereby reprogramming M2 macrophages into M1 macrophages and exposing the core CS(DOX)-PBA to kill MCF-7/ADR cells. Additionally, the nitric oxide (NO) generated by M1 macrophages can suppress the P-glycoprotein (P-gp) expression to reduce the efflux of chemotherapy drugs, thus playing a combined role in overcoming MDR. In vitro studies have demonstrated the effectiveness of GPNP in reprogramming M2 macrophages and inducing apoptosis in MCF-7/ADR cells, resulting in enhanced antitumor efficacy. This work proposed an effective combination strategy to combat chemoresistance, providing new insights into the development of innovative combinatorial therapies against MDR tumors.
Collapse
Affiliation(s)
- Xinzhi Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Chunxiong Zheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China.
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Ying Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Jialei Hao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China.
| |
Collapse
|
56
|
Sun Q, Yang J, Wu Q, Shen W, Yang Y, Yin D. Targeting Lysosome for Enhanced Cancer Photodynamic/Photothermal Therapy in a "One Stone Two Birds" Pattern. ACS APPLIED MATERIALS & INTERFACES 2024; 16:127-141. [PMID: 38118049 DOI: 10.1021/acsami.3c13162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Highly immunogenic programmed death of tumor cells, such as immunogenic cell death (ICD) and pyroptosis, strengthens antitumor responses and thus represents a promising target for cancer immunotherapy. However, the development of ICD and pyroptosis inducers remains challenging, and their efficiency is typically compromised by self-protective autophagy. Here, we report a potent ICD and pyroptosis-inducing strategy by coupling combined photodynamic/photothermal therapy (PTT/PDT) to biological processes in cancer cells. For this purpose, we rationally synthesize a lysosomal-targeting boron-dipyrromethene dimer (BDPd) with intense NIR absorption/emission, high reactive oxygen species (ROS) yield, and photothermal abilities, which can be self-assembled with Pluronic F127, producing lysosomal-acting nanomicelles (BDPd NPs) to facilitate cancer cell internalization of BDPd and generation of intracellular ROS. Owing to the favorable lysosomal-targeting ability of the morpholine group on BDPd, the intracellular BDPd NPs can accumulate in the lysosome and induce robust lysosomal damage in cancer cells upon 660 nm laser irradiation, which results in the synergetic induction of pyroptosis and ICD via activating NLRP3/GSDMD and caspase-3/GSDME pathways simultaneously. More importantly, PTT/PDT-induced self-protective autophagic degradation was blocked due to the dysfunction of lysosomes. Either intratumorally or intravenously, the injected BDPd NPs could markedly inhibit the growth of established tumor tissues upon laser activation, provoke local and systemic antitumor immune responses, and prolong the survival time in the mouse triple-negative breast cancer model. Collectively, this work represents a promising strategy to boost the therapeutic potential of PTT/PDT by coupling phototherapeutic reagents with the subcellular organelles, creating a "one stone two birds" pattern.
Collapse
Affiliation(s)
- Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Jinming Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Qinghua Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021 ,China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021 ,China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
| |
Collapse
|
57
|
Dang BTN, Kwon TK, Lee S, Jeong JH, Yook S. Nanoparticle-based immunoengineering strategies for enhancing cancer immunotherapy. J Control Release 2024; 365:773-800. [PMID: 38081328 DOI: 10.1016/j.jconrel.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/27/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
Cancer immunotherapy is a groundbreaking strategy that has revolutionized the field of oncology compared to other therapeutic strategies, such as surgery, chemotherapy, or radiotherapy. However, cancer complexity, tumor heterogeneity, and immune escape have become the main hurdles to the clinical application of immunotherapy. Moreover, conventional immunotherapies cause many harmful side effects owing to hyperreactivity in patients, long treatment durations and expensive cost. Nanotechnology is considered a transformative approach that enhances the potency of immunotherapy by capitalizing on the superior physicochemical properties of nanocarriers, creating highly targeted tissue delivery systems. These advantageous features include a substantial specific surface area, which enhances the interaction with the immune system. In addition, the capability to finely modify surface chemistry enables the achievement of controlled and sustained release properties. These advances have significantly increased the potential of immunotherapy, making it more powerful than ever before. In this review, we introduce recent nanocarriers for application in cancer immunotherapy based on strategies that target different main immune cells, including T cells, dendritic cells, natural killer cells, and tumor-associated macrophages. We also provide an overview of the role and significance of nanotechnology in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
58
|
Chintalapati SSVV, Iwata S, Miyahara M, Miyako E. Tumor-isolated Cutibacterium acnes as an effective tumor suppressive living drug. Biomed Pharmacother 2024; 170:116041. [PMID: 38113626 DOI: 10.1016/j.biopha.2023.116041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
The two major challenges in cancer treatment are reducing the side effects and minimizing the cost of cancer treatment. A better therapy to treat cancer remains to be developed despite the presence of many therapeutic options. Here, we present bacterial therapy for treating cancer using tumor-isolated Cutibacterium acnes, which is safe to use, has minimal side effects compared to chemotherapeutic drugs, and most importantly, targets the tumor microenvironment due to the bacterium's anaerobic nature. It activates the immune system, and the immune cells effectively penetrate through the tumor tissue and form an immunologic hub inside, explicitly targeting the tumor and destroying the cells. This bacterial therapy is a new cost-effective innovative treatment.
Collapse
Affiliation(s)
| | - Seigo Iwata
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Mikako Miyahara
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Eijiro Miyako
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| |
Collapse
|
59
|
Huang J, Duan F, Xie C, Xu J, Zhang Y, Wang Y, Tang YP, Leung ELH. Microbes mediated immunogenic cell death in cancer immunotherapy. Immunol Rev 2024; 321:128-142. [PMID: 37553793 DOI: 10.1111/imr.13261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/17/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023]
Abstract
Immunogenic cell death (ICD) is one of the 12 distinct cell death forms, which can trigger immune system to fight against cancer cells. During ICD, a number of cellular changes occur that can stimulate an immune response, including the release of molecules called damage-associated molecular patterns (DAMPs), signaling to immune cells to recognize and attack cancer cells. By virtue of their pivotal role in immune surveillance, ICD-based drug development has been a new approach to explore novel therapeutic combinations and personalized strategies in cancer therapy. Several small molecules and microbes can induce ICD-relevant signals and cause cancer cell death. In this review, we highlighted the role of microbe-mediate ICD in cancer immunotherapy and described the mechanisms through which microbes might serve as ICD inducers in cancer treatment. We also discussed current attempts to combine microbes with chemotherapy regimens or immune checkpoint inhibitors (ICIs) in the treatment of cancer patients. We surmise that manipulation of microbes may guide personalized therapeutic interventions to facilitate anticancer immune response.
Collapse
Affiliation(s)
- Jumin Huang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China
| | - Fugang Duan
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, Beijing, China
| | - Chun Xie
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China
| | - Jiahui Xu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China
| | - Yizhong Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Dr. Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Yuwei Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi Province, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi Province, China
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, China
| |
Collapse
|
60
|
Lee S, Kim TD. Breakthroughs in Cancer Immunotherapy: An Overview of T Cell, NK Cell, Mφ, and DC-Based Treatments. Int J Mol Sci 2023; 24:17634. [PMID: 38139461 PMCID: PMC10744055 DOI: 10.3390/ijms242417634] [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: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Efforts to treat cancer using chimeric antigen receptor (CAR)-T therapy have made astonishing progress and clinical trials against hematopoietic malignancies have demonstrated their use. However, there are still disadvantages which need to be addressed: high costs, and side effects such as Graft-versus-Host Disease (GvHD) and Cytokine Release Syndrome (CRS). Therefore, recent efforts have been made to harness the properties of certain immune cells to treat cancer-not just T cells, but also natural killer (NK) cells, macrophages (Mφ), dendritic cells (DC), etc. In this paper, we will introduce immune cell-based cellular therapies that use various immune cells and describe their characteristics and their clinical situation. The development of immune cell-based cancer therapy fully utilizing the unique advantages of each and every immune cell is expected to enhance the survival of tumor patients owing to their high efficiency and fewer side effects.
Collapse
Affiliation(s)
- Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea;
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea;
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
61
|
Zhu MY, Wang T, Wang HD, Wang HZ, Chen HY, Zhang S, Guo YJ, Li H, Hui H. LW-213 induces immunogenic tumor cell death via ER stress mediated by lysosomal TRPML1. Cancer Lett 2023; 577:216435. [PMID: 37806516 DOI: 10.1016/j.canlet.2023.216435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Dying tumor cells release biological signals that exhibit antigenicity, activate cytotoxic T lymphocytes, and induce immunogenic cell death (ICD), playing a key role in immune surveillance. We demonstrate that the flavonoid LW-213 activates endoplasmic reticulum stress (ERS) in different tumor cells and that the lysosomal calcium channel TRPML1 mediates the ERS process in human cellular lymphoma Hut-102 cells. Apoptotic tumor cells induced by ERS often possess immunogenicity. Tumor cells treated with LW-213 exhibit damage-associated molecular patterns (DAMPs), including calreticulin translocation to the plasma membrane and extracellular release of ATP and HMGB1. When co-cultured with antigen-presenting cells (APCs), LW-213-treated tumor cells activated APCs. Two groups of C57BL/6J mice were inoculated with Lewis cells: a "vaccine group", which demonstrated that LW-213-treated tumor cells promote the maturation of dendritic cells and increase CD8+ T cells infiltration in the tumor microenvironment and a "pharmacodynamic group", treated with a combination of LW-213 and PD1/PD-L1 inhibitor (BMS-1), which reduced tumor growth and significantly prolonged the survival time of mice in the "pharmacodynamic group". Therefore, LW-213 can be developed as a novel ICD inducer, providing a new concept for antitumor immunotherapy.
Collapse
Affiliation(s)
- Meng-Yuan Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ting Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hai-di Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hong-Zheng Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hong-Yu Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Shuai Zhang
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yong-Jian Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China.
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China.
| |
Collapse
|
62
|
Ma G, Li F, Wang X, Li Q, Hong Y, Wei Q, Gao F, Zhang W, Guo Y, Ma X, Hu Z. A Bionic Yeast Tumor Vaccine Using the Co-Loading Strategy to Prevent Post-Operative Tumor Recurrence. ACS NANO 2023; 17:21394-21410. [PMID: 37870500 DOI: 10.1021/acsnano.3c06115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Immunotherapy is an effective adjunct to surgery for preventing tumor recurrence and metastasis in postoperative tumor patients. Although mimicking microbial invasion and immune activation pathways can effectively stimulate the immune system, the limited capacity of microbial components to bind antigens and adjuvants restricts the development of this system. Here, we construct bionic yeast carriers (BYCs) by in situ polymerization of mesoporous silica nanoparticles (MSNs) within the yeast capsules (YCs). BYCs can mimic the yeast infection pathway while utilizing the loading capacity of MSNs for multiple substances. Pore size and hydrophobicity-modified BYC can be loaded with both antigen and adjuvant R848. Oral or subcutaneous injection uptake of coloaded BYCs demonstrated positive therapeutic effects as a tumor therapeutic vaccine in both the transplantation tumor model and the metastasis tumor model. 57% of initial 400 mm3 tumor recurrence models are completely cured with coloaded BYCs via combination therapy with surgery, utilizing surgically resected tumors as antigens. The BYCs construction and coloading strategy will provide insights and optimistic approaches for the development of effective and controllable cancer vaccine carriers.
Collapse
Affiliation(s)
- Guanglei Ma
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Fang Li
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Xin Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Qing Li
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang 453003, P. R. China
| | - Youyou Hong
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Qingcong Wei
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Fangli Gao
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Weiwei Zhang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Yuming Guo
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Xiaoming Ma
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Zhiguo Hu
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials. School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| |
Collapse
|
63
|
Ni H, Qian S, Lu J, Feng J, Mou XZ, Zhang J. Natural Polysaccharide Delivery Platforms with Multiscale Structure Used for Cancer Chemoimmunotherapy. Mol Pharm 2023; 20:5778-5789. [PMID: 37752866 DOI: 10.1021/acs.molpharmaceut.3c00633] [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: 09/28/2023]
Abstract
Chemoimmunotherapy is an effective cancer treatment method. Drugs are always combined and used in treating cancer. However, the characteristic of drugs varies, making it challenging to control their release kinetics utilizing delivery devices with a single microstructure. In this study, we attempted to uniformly size drugs of varying molecular weights and confine them in a compartment where immune cells may be recruited and moved freely. Dextran microgels were created as modular drug libraries to address the cryogel burst release of small molecule drugs. Then, modular drug libraries and granulocyte-macrophage colony-stimulating factor (GM-CSF) were integrated into cryogels for a combined treatment. Herein, alginate was zwitterion modified to avoid the immune reaction generated by the material. Because of its macroporous structure, the cryogel could be injected into the body, eliminating invasive surgical procedures. Results demonstrated that multiscale delivery platforms could improve the synergistic effect of various medications on tumor treatment.
Collapse
Affiliation(s)
- Haifeng Ni
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Sunxiang Qian
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Jie Lu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Jie Feng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Xiao-Zhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang 310014, P. R. China
| | - Jing Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| |
Collapse
|
64
|
Li D, Cao Z, Chen C, Li H, He S, Hou X, Liang M, Yang X, Wang J. Nanoassembly of doxorubicin-conjugated polyphosphoester and siRNA simultaneously elicited macrophage- and T cell- mediated anticancer immune response for cancer therapy. Biomaterials 2023; 302:122339. [PMID: 37778054 DOI: 10.1016/j.biomaterials.2023.122339] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/21/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Efficiently reawakening immune cells, including T cells and macrophages, to eliminate tumor cells is a promising strategy for cancer treatment, but remains a huge challenge nowadays. Herein, a nanoassembly formed by doxorubicin (DOX)-conjugated polyphosphoester (PP-(hDOX)) and CD47-targeting siRNA (siCD47) via electrostatic and π-π stacking interactions, termed as PP-(hDOX&siCD47), was developed to reawaken the T cell and macrophage-mediated anticancer activity. The PP-(hDOX&siCD47) could efficiently blockade antiphagocytic signal by downregulation of CD47 expression to reactive macrophage-mediated anticancer immunotherapy. Moreover, the conjugated DOX of PP-(hDOX&siCD47) can perform the chemotherapy towards tumor cells and also elicit the T cell-mediated anticancer immune response via immunogenic cell death (ICD) effect. Therefore, the PP-(hDOX&siCD47) treatment could significantly increase M1-like macrophages proportion and tumor infiltration of CD8+ T cells, while the proportions of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC) were considerably reduced in tumor tissue, eventually achieving significantly tumor growth inhibition. Overall, this study provides a simple siRNA and DOX codelivery approach to simultaneously elicit the macrophage- and T cell-mediated anticancer immune response for cancer therapy.
Collapse
Affiliation(s)
- Dongdong Li
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Ziyang Cao
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Chaoran Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Hengyi Li
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Shan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, PR China
| | - Xurui Hou
- School of International Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, 510006, PR China.
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong, 510006, PR China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong, 510006, PR China.
| |
Collapse
|
65
|
Feng Z, Chen G, Zhong M, Lin L, Mai Z, Tang Y, Chen G, Ma W, Li G, Yang Y, Yu Z, Yu M. An acid-responsive MOF nanomedicine for augmented anti-tumor immunotherapy via a metal ion interference-mediated pyroptotic pathway. Biomaterials 2023; 302:122333. [PMID: 37738743 DOI: 10.1016/j.biomaterials.2023.122333] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Pyroptosis is an inflammatory form of programmed cell death (PCD) that is regulated by the Gasdermin protein family in response to various stimuli, playing a critical role in the development of tumor therapy strategies. However, cancers are generally known to escape from PCD via immunosuppressive pathways or other resistant mechanisms. In this study, an acid-responsive Fe/Mn bimetal-organic framework nanosystem carrying metal ions and immune adjuvant R848 (FeMn@R@H) was designed for combining pyroptosis and augmented immunotherapy. The FeMn@R@H would be triggered to disintegrate and release Fe3+ and Mn2+ ions in response to the acidic tumor microenvironment (TME), thereby initiating Fenton-like reactions for ROS-mediated pyroptosis. On the one hand, the pyroptosis-caused cell rupture would induce the release of proinflammatory cytokines and immunogenic constituents from tumor cells, further resulting in immunogenic cell death (ICD) to promote antitumor immune responses. On the other hand, the co-delivered R848 could reverse suppressive tumor immune microenvironment (TIME) and induce inflammatory responses by activating the TLR7/8 pathway. In conclusion, this tumor-specific therapy system can co-deliver metal ions and R848 to tumor tissues to perform pyroptosis-mediated PCD and augmented anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Zhenzhen Feng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Gui Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Min Zhong
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510632, China
| | - Ling Lin
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Ziyi Mai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Yan Tang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Guimei Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Wen Ma
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China
| | - Guang Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510632, China
| | - Yuanyuan Yang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China.
| | - Zhiqiang Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, China.
| | - Meng Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515 China; Zhujiang Hospital, Southern Medical University, Guangzhou 510282 China.
| |
Collapse
|
66
|
Song Y, Wang R, Pan Y, Fang D, Tian Y, Zhou S. An integrated quorum quenching biocatalytic nanoplatform for synergistic chemo-photothermal eradication of P. aeruginosa biofilm infections. Acta Biomater 2023; 171:532-542. [PMID: 37734627 DOI: 10.1016/j.actbio.2023.09.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023]
Abstract
Decontamination of biofilm-associated infections presents a significant challenge due to the physical and chemical barrier created by the formation of extracellular matrices. This barrier restricts the access of antibiotics to the bacterial communities within the biofilm and provides protection to the persister cells, potentially leading to antibiotic resistance. In this study, we have developed an integrated quorum quenching biocatalytic nanoplatform for the synergistic chemo-photothermal eradication of P. aeruginosa biofilm infections. Ciprofloxacin (Cip), a model antibiotic, was absorbed onto PDA NPs through π-π stacking. Additionally, acylase (AC) was immobilized on PDA NPs through Schiff base reaction and Michael addition, resulting in the formation of the biocatalytic nanoplatform (PDA-Cip-AC NPs). This biocatalytic nanoplatform was able to enzymatically degrade AHL signaling molecules, thus achieving efficient quorum quenching activity to prevent biofilm formation. Furthermore, the NIR light-triggered on-demand Ciprofloxacin release further enhanced the eradication of P. aeruginosa biofilm infections with a synergy of local hyperthermia. We envision that this integrated quorum quenching nanoplatform provides a reliable tool for combating P. aeruginosa biofilm infections. STATEMENT OF SIGNIFICANCE: An integrated quorum quenching biocatalytic nanoplatform has been developed for the eradication of P. aeruginosa biofilm infections. Quorum-sensing signals play a crucial role in modulating bacterial cell-to-cell communication, biofilm formation, and secretion of virulence factors. This biocatalytic nanoplatform efficiently degrades AHL signaling molecules, thereby blocking cell-to-cell communication and preventing biofilm formation. Additionally, local hyperthermia and on-demand Ciprofloxacin release were achieved through NIR irradiation, working synergistically to eradicate P. aeruginosa biofilm infections.
Collapse
Affiliation(s)
- Yue Song
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, PR China
| | - Rui Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, PR China
| | - Yuanzhi Pan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, PR China
| | - Dan Fang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, PR China
| | - Yuan Tian
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, PR China; Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, 518118, PR China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, PR China.
| |
Collapse
|
67
|
Song D, Yang X, Chen Y, Hu P, Zhang Y, Zhang Y, Liang N, Xie J, Qiao L, Deng G, Chen F, Zhang J. Advances in anti-tumor based on various anaerobic bacteria and their derivatives as drug vehicles. Front Bioeng Biotechnol 2023; 11:1286502. [PMID: 37854883 PMCID: PMC10579911 DOI: 10.3389/fbioe.2023.1286502] [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: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
Cancer therapies, such as chemotherapy and radiotherapy, are often unsatisfactory due to several limitations, including drug resistance, inability to cross biological barriers, and toxic side effects on the body. These drawbacks underscore the need for alternative treatments that can overcome these challenges and provide more effective and safer options for cancer patients. In recent years, the use of live bacteria, engineered bacteria, or bacterial derivatives to deliver antitumor drugs to specific tumor sites for controlled release has emerged as a promising therapeutic tool. This approach offers several advantages over traditional cancer therapies, including targeted drug delivery and reduced toxicity to healthy tissues. Ongoing research in this field holds great potential for further developing more efficient and personalized cancer therapies, such as E. coli, Salmonella, Listeria, and bacterial derivatives like outer membrane vesicles (OMVs), which can serve as vehicles for drugs, therapeutic proteins, or antigens. In this review, we describe the advances, challenges, and future directions of research on using live bacteria or OMVs as carriers or components derived from bacteria of delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Daichen Song
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaofan Yang
- School of Clinical Medicine, Jining Medical University, Jining, China
| | - Yanfei Chen
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Pingping Hu
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yingying Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yan Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ning Liang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jian Xie
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lili Qiao
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Guodong Deng
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Fangjie Chen
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiandong Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
68
|
Li Y, Cong Z, Xie L, Tang S, Ren C, Peng X, Tang D, Wan F, Han H, Zhang X, Gao W, Wu S. Magnetically Powered Immunogenic Macrophage Microrobots for Targeted Multimodal Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301489. [PMID: 37300342 DOI: 10.1002/smll.202301489] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/06/2023] [Indexed: 06/12/2023]
Abstract
Motile microrobots open a new realm for disease treatment. However, the concerns of possible immune elimination, targeted capability and limited therapeutic avenue of microrobots constrain its practical biomedical applications. Herein, a biogenic macrophage-based microrobot loaded with magnetic nanoparticles and bioengineered bacterial outer membrane vesicles (OMVs), capable of magnetic propulsion, tumor targeting, and multimodal cancer therapy is reported. Such cell robots preserve intrinsic properties of macrophages for tumor suppression and targeting, and bioengineered OMVs for antitumor immune regulation and fused anticancer peptides. Cell robots display efficient magnetic propulsion and directional migration in the confined space. In vivo tests show that cell robots can accumulate at the tumor site upon magnetic manipulation, coupling with tumor tropism of macrophages to greatly improve the efficacy of its multimodal therapy, including tumor inhibition of macrophages, immune stimulation, and antitumor peptides of OMVs. This technology offers an attractive avenue to design intelligent medical microrobots with remote manipulation and multifunctional therapy capabilities for practical precision treatment.
Collapse
Affiliation(s)
- Yangyang Li
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Zhaoqing Cong
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Leiming Xie
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Songsong Tang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Chunyu Ren
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Xiqi Peng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Daitian Tang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Fangchen Wan
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
| | - Hong Han
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xueji Zhang
- School of Biomedical Engineering, Health Science Centre, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Song Wu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, P. R. China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518000, P. R. China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, P. R. China
| |
Collapse
|
69
|
Ya Z, Guo S, Li Y, Zhu M, Zhang L, Zong Y, Wan M. Focused acoustic vortex-mediated sonochemotherapy for the amplification of immunogenic cell death combined with checkpoint blockade to potentiate cancer immunotherapy. Biomaterials 2023; 301:122278. [PMID: 37598439 DOI: 10.1016/j.biomaterials.2023.122278] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/03/2023] [Accepted: 08/12/2023] [Indexed: 08/22/2023]
Abstract
Sonodynamic therapy (SDT) as an auxiliary modality of cancer immunotherapy enhances systemic anti-tumor immunity. However, the efficiency of SDT-mediated immunotherapy based on conventional focused ultrasound (FUS) is restricted by the tiny focal region of FUS. Focused acoustic vortex (FAV) possessing a larger focal region, can induce stronger cavitation and thermal effects than FUS with the same parameters, having the potential to overcome this issue. This research investigated the feasibility of FAV-mediated sonochemotherapy combined with the immune checkpoint blockade (ICB) to reshape immunosuppressive tumor microenvironment (TME), inhibit tumor growth and lung metastasis. Sonosensitizer chlorin e6 (Ce6) and chemotherapeutic agent doxorubicin (Dox) were co-loaded into microbubble-liposome complex to compose Ce6/Dox@Lip@MBs (CDLM) for "all-in-one" synergistic sonochemotherapy, whose main components were clinical approved. FAV-activated CDLM significantly enriched immunogenic cell death (ICD) inducers in tumors and amplified ICD of cancer cells compared with FUS-activated CDLM. Furthermore, the amplified-ICD combined with ICB increased the infiltration of cytotoxic T lymphocytes and natural killer cells, polarized M2 macrophages to M1 macrophages, and decreased regulatory T cells. This study provides a multifunctional strategy for enriching ICD inducers in tumors and amplifying ICD to ameliorate immunosuppressive TME and potentiate systemic anti-tumor immunity.
Collapse
Affiliation(s)
- Zhen Ya
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shifang Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yan Li
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Mingting Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Lei Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yujin Zong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| | - Mingxi Wan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| |
Collapse
|
70
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
71
|
Qin Y, Zhang H, Li Y, Xie T, Yan S, Wang J, Qu J, Ouyang F, Lv S, Guo Z, Wei H, Yu CY. Promotion of ICD via Nanotechnology. Macromol Biosci 2023; 23:e2300093. [PMID: 37114599 DOI: 10.1002/mabi.202300093] [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: 03/07/2023] [Revised: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy represents the most promising treatment strategy for cancer, but suffers from compromised therapeutic efficiency due to low immune activity of tumor cells and an immunosuppressive microenvironment, which significantly hampers the clinical translations of this treatment strategy. To promote immunotherapy with desired therapeutic efficiency, immunogenic cell death (ICD), a particular type of death capable of reshaping body's antitumor immune activity, has drawn considerable attention due to the potential to stimulate a potent immune response. Still, the potential of ICD effect remains unsatisfactory because of the intricate tumor microenvironment and multiple drawbacks of the used inducing agents. ICD has been thoroughly reviewed so far with a general classification of ICD as a kind of immunotherapy strategy and repeated discussion of the related mechanism. However, there are no published reviews, to the authors' knowledge, providing a systematic summarization on the enhancement of ICD via nanotechnology. For this purpose, this review first discusses the four stages of ICD according to the development mechanisms, followed by a comprehensive description on the use of nanotechnology to enhance ICD in the corresponding four stages. The challenges of ICD inducers and possible solutions are finally summarized for future ICD-based enhanced immunotherapy.
Collapse
Affiliation(s)
- Yang Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxian Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ting Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shuang Yan
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaqi Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jun Qu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Feijun Ouyang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shaoyang Lv
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zifen Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
72
|
Jiang X, Wu L, Zhang M, Zhang T, Chen C, Wu Y, Yin C, Gao J. Biomembrane nanostructures: Multifunctional platform to enhance tumor chemoimmunotherapy via effective drug delivery. J Control Release 2023; 361:510-533. [PMID: 37567505 DOI: 10.1016/j.jconrel.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Chemotherapeutic drugs have been found to activate the immune response against tumors by inducing immunogenic cell death, in addition to their direct cytotoxic effects toward tumors, therefore broadening the application of chemotherapy in tumor immunotherapy. The combination of other therapeutic strategies, such as phototherapy or radiotherapy, could further strengthen the therapeutic effects of immunotherapy. Nanostructures can facilitate multimodal tumor therapy by integrating various active agents and combining multiple types of therapeutics in a single nanostructure. Biomembrane nanostructures (e.g., exosomes and cell membrane-derived nanostructures), characterized by superior biocompatibility, intrinsic targeting ability, intelligent responsiveness and immune-modulating properties, could realize superior chemoimmunotherapy and represent next-generation nanostructures for tumor immunotherapy. This review summarizes recent advances in biomembrane nanostructures in tumor chemoimmunotherapy and highlights different types of engineering approaches and therapeutic mechanisms. A series of engineering strategies for combining different biomembrane nanostructures, including liposomes, exosomes, cell membranes and bacterial membranes, are summarized. The combination strategy can greatly enhance the targeting, intelligence and functionality of biomembrane nanostructures for chemoimmunotherapy, thereby serving as a stronger tumor therapeutic method. The challenges associated with the clinical translation of biomembrane nanostructures for chemoimmunotherapy and their future perspectives are also discussed.
Collapse
Affiliation(s)
- Xianghe Jiang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Cuimin Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China.
| | - Chuan Yin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
73
|
Zheng J, Jiang J, Pu Y, Xu T, Sun J, Zhang Q, He L, Liang X. Tumor-associated macrophages in nanomaterial-based anti-tumor therapy: as target spots or delivery platforms. Front Bioeng Biotechnol 2023; 11:1248421. [PMID: 37654704 PMCID: PMC10466823 DOI: 10.3389/fbioe.2023.1248421] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Targeting tumor-associated macrophages (TAMs) has emerged as a promising approach in cancer therapy. This article provides a comprehensive review of recent advancements in the field of nanomedicines targeting TAMs. According to the crucial role of TAMs in tumor progression, strategies to inhibit macrophage recruitment, suppress TAM survival, and transform TAM phenotypes are discussed as potential therapeutic avenues. To enhance the targeting capacity of nanomedicines, various approaches such as the use of ligands, immunoglobulins, and short peptides are explored. The utilization of live programmed macrophages, macrophage cell membrane-coated nanoparticles and macrophage-derived extracellular vesicles as drug delivery platforms is also highlighted, offering improved biocompatibility and prolonged circulation time. However, challenges remain in achieving precise targeting and controlled drug release. The heterogeneity of TAMs and the variability of surface markers pose hurdles in achieving specific recognition. Furthermore, the safety and clinical applicability of these nanomedicines requires further investigation. In conclusion, nanomedicines targeting TAMs hold great promise in cancer therapy, offering enhanced specificity and reduced side effects. Addressing the existing limitations and expanding our understanding of TAM biology will pave the way for the successful translation of these nano-therapies into clinical practice.
Collapse
Affiliation(s)
- Jixuan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jinting Jiang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yicheng Pu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Tingrui Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jiantong Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qiang Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Liang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
74
|
Zhou M, Tang Y, Xu W, Hao X, Li Y, Huang S, Xiang D, Wu J. Bacteria-based immunotherapy for cancer: a systematic review of preclinical studies. Front Immunol 2023; 14:1140463. [PMID: 37600773 PMCID: PMC10436994 DOI: 10.3389/fimmu.2023.1140463] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/30/2023] [Indexed: 08/22/2023] Open
Abstract
Immunotherapy has been emerging as a powerful strategy for cancer management. Recently, accumulating evidence has demonstrated that bacteria-based immunotherapy including naive bacteria, bacterial components, and bacterial derivatives, can modulate immune response via various cellular and molecular pathways. The key mechanisms of bacterial antitumor immunity include inducing immune cells to kill tumor cells directly or reverse the immunosuppressive microenvironment. Currently, bacterial antigens synthesized as vaccine candidates by bioengineering technology are novel antitumor immunotherapy. Especially the combination therapy of bacterial vaccine with conventional therapies may further achieve enhanced therapeutic benefits against cancers. However, the clinical translation of bacteria-based immunotherapy is limited for biosafety concerns and non-uniform production standards. In this review, we aim to summarize immunotherapy strategies based on advanced bacterial therapeutics and discuss their potential for cancer management, we will also propose approaches for optimizing bacteria-based immunotherapy for facilitating clinical translation.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yucheng Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyan Hao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yongjiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Si Huang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| |
Collapse
|
75
|
Wang Y, Li S, Ren X, Yu S, Meng X. Nano-engineering nanomedicines with customized functions for tumor treatment applications. J Nanobiotechnology 2023; 21:250. [PMID: 37533106 PMCID: PMC10399036 DOI: 10.1186/s12951-023-01975-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 06/29/2023] [Indexed: 08/04/2023] Open
Abstract
Nano-engineering with unique "custom function" capability has shown great potential in solving technical difficulties of nanomaterials in tumor treatment. Through tuning the size and surface properties controllablly, nanoparticles can be endoewd with tailored structure, and then the characteristic functions to improve the therapeutic effect of nanomedicines. Based on nano-engineering, many have been carried out to advance nano-engineering nanomedicine. In this review, the main research related to cancer therapy attached to the development of nanoengineering nanomedicines has been presented as follows. Firstly, therapeutic agents that target to tumor area can exert the therapeutic effect effectively. Secondly, drug resistance of tumor cells can be overcome to enhance the efficacy. Thirdly, remodeling the immunosuppressive microenvironment makes the therapeutic agents work with the autoimmune system to eliminate the primary tumor and then prevent tumor recurrence and metastasis. Finally, the development prospects of nano-engineering nanomedicine are also outlined.
Collapse
Affiliation(s)
- Yuxin Wang
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shimei Li
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiangling Ren
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Shiping Yu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China.
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
76
|
Liang Y, Wang PY, Liu ZY, Sun HF, Wang Q, Sun GB, Zhang X, Li YJ, Xie SY. Dual Stimuli-Responsive Micelles for Imaging-Guided Mitochondrion-Targeted Photothermal/Photodynamic/Chemo Combination Therapy-Induced Immunogenic Cell Death. Int J Nanomedicine 2023; 18:4381-4402. [PMID: 37551273 PMCID: PMC10404442 DOI: 10.2147/ijn.s410047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/14/2023] [Indexed: 08/09/2023] Open
Abstract
Introduction As the special modality of cell death, immunogenic cell death (ICD) could activate immune response. Phototherapy in combination with chemotherapy (CT) is a particularly efficient tumor ICD inducing method that could overcome the defects of monotherapies. Methods In this study, new dual stimuli-responsive micelles were designed and prepared for imaging-guided mitochondrion-targeted photothermal/photodynamic/CT combination therapy through inducing ICD. A dual-sensitive methoxy-polyethylene glycol-SS-poly(L-γ-glutamylglutamine)-SS-IR780 (mPEG-SS-PGG-SS-IR780) polymer was synthesized by grafting IR780 with biodegradable di-carboxyl PGG as the backbone, and mPEG-SS-PGG-SS-IR780/paclitaxel micelles (mPEG-SS-PGG-SS-IR780/PTXL MCs) were synthesized by encapsulating PTXL in the hydrophobic core. Results In-vivo and -vitro results demonstrated that the three-mode combination micelles inhibited tumor growth and enhanced the therapeutic efficacy of immunotherapy. The dual stimuli-responsive mPEG-SS-PGG-SS-IR780/PTXL MCs were able to facilitate tumor cell endocytosis of nanoparticles. They were also capable of promoting micelles disintegration and accelerating PTXL release. The mPEG-SS-PGG-SS-IR780/PTXL MCs induced mitochondrial dysfunction by directly targeting the mitochondria, considering the thermo- and reactive oxygen species (ROS) sensitivity of the mitochondria. Furthermore, the mPEG-SS-PGG-SS-IR780/PTXL MCs could play the diagnostic and therapeutic roles via imaging capabilities. Conclusion In summary, this study formulated a high-efficiency nanoscale platform with great potential in combined therapy for tumors through ICD.
Collapse
Affiliation(s)
- Yan Liang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, QingDao, ShanDong, 266071, People’s Republic of China
| | - Ping-Yu Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Ze-Yun Liu
- School of International Studies, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hong-Fang Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Qin Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Guang-Bin Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Xia Zhang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - You-Jie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Shu-Yang Xie
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, QingDao, ShanDong, 266071, People’s Republic of China
| |
Collapse
|
77
|
Yoon J, Le XT, Kim J, Lee H, Nguyen NT, Lee WT, Lee ES, Oh KT, Choi HG, Youn YS. Macrophage-reprogramming upconverting nanoparticles for enhanced TAM-mediated antitumor therapy of hypoxic breast cancer. J Control Release 2023; 360:482-495. [PMID: 37423526 DOI: 10.1016/j.jconrel.2023.07.009] [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: 02/26/2023] [Revised: 06/10/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
In an attempt to achieve antitumor effects by switching the phenotype of macrophages from the tumor-promoting M2 type to the tumor-suppressing M1 type, we fabricated mannose-decorated/macrophage membrane-coated, silica-layered NaErF4@NaLuF4 upconverting nanoparticles (UCNPs) co-doped with perfluorocarbon (PFC)/chlorin e6 (Ce6) and loaded with paclitaxel (PTX) (UCNP@mSiO2-PFC/Ce6@RAW-Man/PTX: ∼61 nm; -11.6 mV). These nanoparticles were designed to have two major functionalities, (i) efficient singlet oxygen generation aided by an oxygen supply and (ii) good targeting to tumor-associated macrophage (TAMs) (M2-type), to induce polarization to M1 type macrophages that release proinflammatory cytokines and suppress breast cancers. The primary UCNPs consisted of lanthanide elements (erbium and lutetium) in a core@shell structure, and they facilely emitted 660 nm light in response to a deep-penetrating 808 nm near-infrared laser. Moreover, the UCNPs@mSiO2-PFC/Ce6@RAW-Man/PTX were able to release O2 and generate 1O2 because of the co-doped PFC/Ce6 and upconversion. Our nanocarriers' excellent uptake to RAW 264.7 macrophage cells (M2 type) and efficient M1-type polarization activity were clearly demonstrated using qRT-PCR and immunofluorescence-based confocal laser scanning microscopy. Our nanocarriers displayed significant cytotoxicity to 4T1 cells in 2D culture and 3D co-culture systems of 4T1/RAW 264.7 cells. More importantly, UCNPs@mSiO2-PFC/Ce6@RAW-Man/PTX (+808 nm laser) noticeably suppressed tumor growth in 4T1-xenografted mice, compared with the other treatment groups (332.4 vs. 709.5-1185.5 mm3). We attribute this antitumor efficacy to the prominent M1-type macrophage polarization caused by our nanocarriers through efficient ROS/O2 generation and targeting of M2-type TAMs via mannose ligands on coated macrophage-membrane.
Collapse
Affiliation(s)
- Johyun Yoon
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Juho Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hyunjun Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Eun Seong Lee
- Department of Biotechnology and Department of Biomedical-Chemical Engineering, Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
78
|
Zhang H, Wang Y, Zhu L, Qi Z, Cao K, Chang J, Hou L. A "bulldozer" driven by anoxic bacteria for pancreatic cancer chemo-immunotherapy. J Control Release 2023; 360:660-671. [PMID: 37433371 DOI: 10.1016/j.jconrel.2023.07.014] [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: 04/24/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023]
Abstract
Immune evasion is a major obstacle for pancreatic ductal adenocarcinoma (PDAC) therapy. Inhibition of autophagy can improve antigen presentation and enlarge immunogenic cell death (ICD) effect to generate a strong anti-tumor immune response. However, abundant extracellular matrix dominated by hyaluronic acid (HA) hinders the deep penetration of autophagy inhibitors and ICD inducers. Herein, an intelligent autophagy inhibitor hydroxychloroquine (HCQ) and chemotherapeutic drug doxorubicin (DOX) co-loaded "bulldozer" (HD@HH/EcN) driven by anoxic bacteria was constructed for PDAC chemo-immunotherapy. Results demonstrated that probiotic Escherichia coli 1917 (EcN) could carry hyaluronidases (HAases)-hybrided albumin nanoparticles (HD@HH) to reach PDAC tumor tissue quickly and accurately. Thereafter, HAases can efficiently cleave the tumor matrix barrier and promote HD@HH/EcN to accumulate at tumor hypoxic core significantly. After that, high level of glutathione (GSH) in tumor microenvironment (TME) induces intermolecular disulfide bond in HD@HH nanoparticles breakage, to precisely release HCQ and DOX. DOX can induce ICD effect. Meanwhile, HCQ can amplify DOX induced ICD effect by inhibiting tumor autophagy, which further increase cell surface expression of major histocompatibility complex class I (MHC-I) and augment recruitment of CD8+ T cell to improve immunosuppressive TME. This study provides a new strategy for PDAC chemo-immunotherapy.
Collapse
Affiliation(s)
- Huijuan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China.
| | - Yaping Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ling Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China
| | - Zijun Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Kexuan Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junbiao Chang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China; School of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China.
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China.
| |
Collapse
|
79
|
Huang C, Xie T, Liu Y, Yan S, OuYang F, Zhang H, Lei L, He D, Wei H, Yu CY. A Sodium Alginate-Based Multifunctional Nanoplatform for Synergistic Chemo-Immunotherapy of Hepatocellular Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301352. [PMID: 37216573 DOI: 10.1002/adma.202301352] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/13/2023] [Indexed: 05/24/2023]
Abstract
Efficient hepatocellular carcinoma (HCC) treatment remains a significant challenge due to the inherent limitations of traditional strategies. The exploration of polysaccharides' natural immunity for HCC immunotherapy is rarely explored. For this purpose, facile construction of a multifunctional nanoplatform, biotinylated aldehyde alginate-doxorubicin nano micelle (BEACNDOXM) is reported in this study for synergistic chemo-immunotherapy by using constant β-D-mannuronic acid (M) units and modulated α-L-guluronic acid (G) units in the alginate (ALG) structure. The M units show natural immunity and specific binding ability with mannose receptors (MRs) via strong receptor-ligand interactions, and the G units serve as highly reactive conjugation sites for biotin (Bio) and DOX. Therefore, this formulation not only integrates the natural immunity of ALG and the immunogenic cell death (ICD) triggering function of DOX, but also shows dual targeting properties to HCC cells via MRs and Bio receptors (BRs)-mediated endocytosis. Notably, BEACNDOXM mediates a tumor inhibitory efficiency 12.10% and 4.70% higher than free DOX and single targeting aldehyde alginate-doxorubicin nano micelle controls, respectively, at an equivalent DOX dose of 3 mg kg-1 in Hepa1-6 tumor-bearing mice. This study reports the first example of integrating the natural immunity of ALG and the ICD effect of anticancer drugs for enhanced chemo-immunotherapy of HCC.
Collapse
Affiliation(s)
- Cong Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Ting Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Yufeng Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Shuang Yan
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Feijun OuYang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Longtianyang Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Dongxiu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
80
|
Shahin RK, Elkady MA, Abulsoud AI, Abdelmaksoud NM, Abdel Mageed SS, El-Dakroury WA, Zewail MB, Elazazy M, Sobhy MH, Nomier Y, Elazazy O, Elballal MS, Mohammed OA, Midan HM, Elrebehy MA, Ziada BO, Doghish AS. miRNAs orchestration of gallbladder cancer - Particular emphasis on diagnosis, progression and drug resistance. Pathol Res Pract 2023; 248:154684. [PMID: 37454489 DOI: 10.1016/j.prp.2023.154684] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Gallbladder cancer (GBC) is characterized by a highly invasive nature and a poor prognosis, with adenocarcinoma being the main histological subtype. According to statistical data, patients diagnosed with advanced GBC have a survival rate of less than 5% for 5 years. Despite the novel therapeutic techniques, the unsatisfactory results could be related to the underlying biology of tumor cells and resistance to chemotherapy. Early diagnosis is more important than clinical therapy as it assists in determining the pathological stage of cancer and facilitates the selection of appropriate medication. Hence, it is very important to understand the precise pathogenesis of GBC and to discover potential novel biomarkers for early diagnosis of GBC. Non-coding RNAs, such as microRNAs, long non-coding RNAs, and circular RNAs, have been found to influence the transcriptional regulation of target genes associated with cancer, either directly or indirectly. microRNAs are a group of small, non-coding, single-stranded RNAs that are expressed endogenously. miRNAs play significant roles in various fundamental cellular processes. Therefore, miRNAs have the potential to serve as valuable biomarkers and therapeutic targets for GBC.
Collapse
Affiliation(s)
- Reem K Shahin
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohamed A Elkady
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | | | - Sherif S Abdel Mageed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Moataz B Zewail
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud Elazazy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed H Sobhy
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, Egypt
| | - Yousra Nomier
- Pharmacology Department, Pharmacy College, Jazan University, Saudi Arabia
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, Faculty of Medicine, Bisha University, Bisha 61922, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Heba M Midan
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Bassant O Ziada
- Research Department, Utopia Pharmaceuticals, Nasr City, 11765 Cairo, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
81
|
Song P, Han X, Li X, Cong Y, Wu Y, Yan J, Wang Y, Wang X, Mu Z, Wang L, Li X, Zhang H. Bacteria engineered with intracellular and extracellular nanomaterials for hierarchical modulation of antitumor immune responses. MATERIALS HORIZONS 2023; 10:2927-2935. [PMID: 37158992 DOI: 10.1039/d3mh00249g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Induction of immunogenic cell death (ICD) by hyperthermia can initiate adaptive immune responses, emerging as an attractive strategy for tumor immunotherapy. However, ICD can induce proinflammatory factor interferon-γ (IFN-γ) production, leading to indoleamine 2,3-dioxygenase 1 (IDO-1) activation and an immunosuppressive tumor microenvironment, which dramatically reduces the ICD-triggered immunotherapeutic efficacy. Herein, we developed a bacteria-nanomaterial hybrid system (CuSVNP20009NB) to systematically modulate the tumor immune microenvironment and improve tumor immunotherapy. Attenuated Salmonella typhimurium (VNP20009) that can chemotactically migrate to the hypoxic area of the tumor and repolarize tumor-associated macrophages (TAMs) was employed to intracellularly biosynthesize copper sulfide nanomaterials (CuS NMs) and extracellularly hitchhike NLG919-embedded and glutathione (GSH)-responsive albumin nanoparticles (NB NPs), forming CuSVNP20009NB. After intravenous injection into B16F1 tumor-bearing mice, CuSVNP20009NB could accumulate in tumor tissues and repolarize TAMs from the immunosuppressive M2 to immunostimulatory M1 phenotype and release NLG919 from extracellular NB NPs to inhibit IDO-1 activity. Under further near infrared laser irradiation, intracellular CuS NMs of CuSVNP20009NB could photothermally induce ICD including calreticulin (CRT) expression and high mobility group box 1 (HMGB-1) release, promoting intratumoral infiltration of cytotoxic T lymphocytes. Finally, CuSVNP20009NB with excellent biocompatibility could systematically augment immune responses and significantly inhibit tumor growth, holding great promise for tumor therapy.
Collapse
Affiliation(s)
- Panpan Song
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
- University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Xiaoqing Han
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
| | - Xiumin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, China
| | - Yalin Cong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, China
| | - Yunyun Wu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, 130012, China.
| | - Jiao Yan
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
| | - Yanjing Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
- University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Xingbo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
- University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Zhengzhi Mu
- Key Laboratory of Bionic Engineering (Ministry of Education, China), Jilin University, Changchun, 130022, China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Xi Li
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, 130012, China.
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
- University of Science and Technology of China, Hefei, 230026, Anhui, China
| |
Collapse
|
82
|
Park SH, Eun R, Heo J, Lim YT. Nanoengineered drug delivery in cancer immunotherapy for overcoming immunosuppressive tumor microenvironment. Drug Deliv Transl Res 2023; 13:2015-2031. [PMID: 36581707 DOI: 10.1007/s13346-022-01282-8] [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] [Accepted: 12/08/2022] [Indexed: 12/31/2022]
Abstract
Almost like a living being in and of itself, tumors actively interact with and modify their environment to escape immune responses. Owing to the pre-formation of cancer-favorable microenvironment prior to anti-cancer treatment, the numerous attempts that followed propose limited efficacy in oncology. Immunogenicity by activation of immune cells within the tumor microenvironment or recruitment of immune cells from nearby lymph nodes is quickly offset as the immunosuppressive environment, rapidly converting immunogenic cells into immune suppressive cells, overriding the immune system. Tumor cells, as well as regulatory cells, namely M2 macrophages, Treg cells, and MDSCs, derived by the immunosuppressive environment, also cloak from potential anti-tumoral factors by directly or indirectly secreting cytokines, such as IL-10 and TGF-β, related to immune regulation. Enzymes and other metabolic or angiogenetic constituents - VEGF, IDO1, and iNOS - are also employed directed for anti-cancer immune cell malfunctioning. Therefore, the conversion of "cold" immunosuppressive environment into "hot" immune responsive environment is of paramount importance, bestowing the advances in the field of cancer immunotherapy the opportunity to wholly fulfill its intended purpose. This paper reviews the mechanisms by which tumors wield to exercise immune suppression and the nanoengineered delivery strategies being developed to overcome this suppression.
Collapse
Affiliation(s)
- Sei Hyun Park
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Ryounho Eun
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Janghun Heo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea.
| |
Collapse
|
83
|
Gong T, Wu J. Synthetic engineered bacteria for cancer therapy. Expert Opin Drug Deliv 2023; 20:993-1013. [PMID: 37497622 DOI: 10.1080/17425247.2023.2241367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/10/2023] [Accepted: 07/24/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Cancer mortality worldwide highlights the urgency for advanced therapeutic methods to fill the gaps in conventional cancer therapies. Bacteriotherapy is showing great potential in tumor regression due to the motility and colonization tendencies of bacteria. However, the complicated in vivo environment and tumor pathogenesis hamper the therapeutic outcomes. Synthetic engineering methods endow bacteria with flexible abilities both at the extracellular and intracellular levels to meet treatment requirements. In this review, we introduce synthetic engineering methods for bacterial modifications. We highlight the recent progress in engineered bacteria and explore how these synthetic methods endow bacteria with superior abilities in cancer therapy. The current clinical translations are further discussed. Overall, this review may shed light on the advancement of engineered bacteria for cancer therapy. AREAS COVERED Recent progress in synthetic methods for bacterial engineering and specific examples of their applications in cancer therapy are discussed in this review. EXPERT OPINION Bacteriotherapy bridges the gaps of conventional cancer therapies through the natural motility and colonization tendency of bacteria, as well as their synthetic engineering. Nevertheless, to fulfill the bacteriotherapy potential and move into clinical trials, more research focusing on its safety concerns should be conducted.
Collapse
Affiliation(s)
- Tong Gong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| |
Collapse
|
84
|
Narmani A, Jahedi R, Bakhshian-Dehkordi E, Ganji S, Nemati M, Ghahramani-Asl R, Moloudi K, Hosseini SM, Bagheri H, Kesharwani P, Khani A, Farhood B, Sahebkar A. Biomedical applications of PLGA nanoparticles in nanomedicine: advances in drug delivery systems and cancer therapy. Expert Opin Drug Deliv 2023; 20:937-954. [PMID: 37294853 DOI: 10.1080/17425247.2023.2223941] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/06/2023] [Indexed: 06/11/2023]
Abstract
INTRODUCTION During the last decades, the ever-increasing proportion of patients with cancer has been led to serious concerns worldwide. Therefore, the development and use of novel pharmaceuticals, like nanoparticles (NPs)-based drug delivery systems (DDSs), can be potentially effective in cancer therapy. AREA COVERED Poly lactic-co-glycolic acid (PLGA) NPs, as a kind of bioavailable, biocompatible, and biodegradable polymers, have approved by the Food and Drug Administration (FDA) for some biomedical and pharmaceutical applications. PLGA is comprised of lactic acid (LA) and glycolic acid (GA) and their ratio could be controlled during various syntheses and preparation approaches. LA/GA ratio determines the stability and degradation time of PLGA; lower content of GA results in fast degradation. There are several approaches for preparing PLGA NPs that can affect their various aspects, such as size, solubility, stability, drug loading, pharmacokinetics, and pharmacodynamics, and so on. EXPERT OPINION These NPs have indicated the controlled and sustained drug release in the cancer site and can use in passive and active (via surface modification) DDSs. This review aims to provide an overview of PLGA NPs, their preparation approach and physicochemical aspects, drug release mechanism and the cellular fate, DDSs for efficient cancer therapy, and status in the pharmaceutical industry and nanomedicine.
Collapse
Affiliation(s)
- Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Roghayyeh Jahedi
- Department of Plant Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Ehsan Bakhshian-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Saeid Ganji
- Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ruhollah Ghahramani-Asl
- Department of Medical Physics and Radiological Sciences, Faculty of Paramedicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Kave Moloudi
- Department of Radiology and Nuclear Medicine, Alley School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Mohammad Hosseini
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Ali Khani
- Radiation Sciences Department, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
85
|
Hu T, Huang Y, Liu J, Shen C, Wu F, He Z. Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy. Pharmaceutics 2023; 15:1821. [PMID: 37514008 PMCID: PMC10383408 DOI: 10.3390/pharmaceutics15071821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy can significantly prevent tumor growth and metastasis by activating the autoimmune system without destroying normal cells. Although cancer immunotherapy has made some achievements in clinical cancer treatment, it is still restricted by systemic immunotoxicity, immune cell dysfunction, cancer heterogeneity, and the immunosuppressive tumor microenvironment (ITME). Biomimetic cell-derived nanoparticles are attracting considerable interest due to their better biocompatibility and lower immunogenicity. Moreover, biomimetic cell-derived nanoparticles can achieve different preferred biological effects due to their inherent abundant source cell-relevant functions. This review summarizes the latest developments in biomimetic cell-derived nanoparticles for cancer immunotherapy, discusses the applications of each biomimetic system in cancer immunotherapy, and analyzes the challenges for clinical transformation.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuezhou Huang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chao Shen
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fengbo Wu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
86
|
Li H, Yang X, Wang Z, She W, Liu Y, Huang L, Jiang P. A Near-Infrared-II Fluorescent Nanocatalyst for Enhanced CAR T Cell Therapy against Solid Tumor by Immune Reprogramming. ACS NANO 2023. [PMID: 37319120 DOI: 10.1021/acsnano.3c02592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy holds great promise in the treatment of hematological malignancies but performs poorly in solid tumors due to the tumor immunosuppressive microenvironment. Herein, a multifunctional nanocatalyst (APHA@CM) was prepared by encapsulating horseradish peroxidase (HRP)-loaded Au/polydopamine nanoparticles (Au/PDA NPs) and Ag2S quantum dots with CAR T cell membranes to improve the CAR T cell therapy in solid tumors. The APHA@CM has excellent multimodal imaging capability to precisely guide the scope and time window for nanocatalyst-induced tumor microenvironment regulation and CAR T cell therapy. The oxidase-like activity of Au NPs inhibited the glycolytic metabolism of tumor cells, reducing lactate efflux, reprogramming tumor immunosuppression, and ultimately increasing CAR T cell activation within the tumors. Additionally, the hypoxia environment of tumors could be relieved by HRP to enhance the Au/PDA NPs-induced synergistic sonodynamic/photothermal therapy (SDT/PTT), thereby promoting the immunogenic cell death of NALM 6 cells and enhancing CAR T cell-mediated immune microenvironment reprogramming. When this strategy was utilized to treat NALM 6 solid tumors, it not only completely eliminated tumors but also formed a long-term immune memory effect to inhibit tumor metastasis and recurrence. This work offers a strategy for CAR T cell therapy in solid tumor.
Collapse
Affiliation(s)
- Haimei Li
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| | - Xiuxiu Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zichen Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Wenyan She
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yi Liu
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Green Chemical Technology and Process Engineering, School of Chemistry and Chemical Engineering, Tiangong University, Tianjin 300387, China
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, Xianning 437100, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan 430030, China
| | - Peng Jiang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| |
Collapse
|
87
|
Huang G, Liu L, Pan H, Cai L. Biomimetic Active Materials Guided Immunogenic Cell Death for Enhanced Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201412. [PMID: 36572642 DOI: 10.1002/smtd.202201412] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/22/2022] [Indexed: 05/17/2023]
Abstract
Despite immunotherapy emerging as a vital approach to improve cancer treatment, the activation of efficient immune responses is still hampered by immunosuppression, especially due to the low tumor immunogenicity. Immunogenic cell death (ICD) is a promising strategy to reshape the tumor microenvironment (TME) for achieving high immunogenicity. Various stimuli are able to effectively initiate their specific ICD by utilizing the corresponding ICD-inducer. However, the ICD-guided antitumor immune effects are usually impaired by various biological barriers and TME-associated immune resistance. Biomimetic active materials are being extensively explored as guided agents for ICD due to their unique advantages. In this review, two major strategies are systematically introduced that have been employed to exploit biomimetic active materials guided ICD for cancer immunotherapy, mainly including naive organism-derived nanoagents and engineered bioactive platforms. This review outlines the recent advances in the field at biomimetic active materials guided physiotherapy, chemotherapy, and biotherapy for ICD induction. The advances and challenges of biomimetic active materials guided ICD for cancer immunotherapy applications are further discussed in future clinical practice. This review provides an overview of the advances of biomimetic active materials for targeting immunoregulation and treatment and can contribute to the future of advanced antitumor combination therapy.
Collapse
Affiliation(s)
- Guojun Huang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
88
|
Shen S, Chen Y, Zhou J, Zhang H, Xia X, Yang Y, Zhang Y, Noori A, Mousavi MF, Chen M, Xia Y, Zhang W. Microbe‐Mediated Biosynthesis of Multidimensional Carbon‐Based Materials for Energy Storage Applications. ADVANCED ENERGY MATERIALS 2023; 13. [DOI: 10.1002/aenm.202204259] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 01/06/2025]
Abstract
AbstractBiosynthesis methods are considered to be a promising technology for engineering new carbon‐based materials or redesigning the existing ones for specific purposes with the aid of synthetic biology. Lots of biosynthetic processes including metabolism, fermentation, biological mineralization, and gene editing have been adopted to prepare novel carbon‐based materials with exceptional properties that cannot be realized by traditional chemical methods, because microbes evolved to possess special abilities to modulate components/structure of materials. In this review, the recent development on carbon‐based materials prepared via different biosynthesis methods and various microbe factories (such as bacteria, yeasts, fungus, viruses, proteins) are systematically reviewed. The types of biotechniques and the corresponding mechanisms for the synthesis of carbon‐based materials are outlined. This review also focuses on the structural design and compositional engineering of carbon‐based nanostructures (e.g., metals, semiconductors, metal oxides, metal sulfides, phosphates, Mxenes) derived from biotechnology and their applications in electrochemical energy storage devices. Moreover, the relationship of the architecture–composition–electrochemical behavior and performance enhancement mechanism is also deeply discussed and analyzed. Finally, the development perspectives and challenges on the biosynthetic carbons are proposed and may pave a new avenue for rational design of advanced materials for the low‐carbon economy.
Collapse
Affiliation(s)
- Shenghui Shen
- School of Materials Science and Engineering Zhejiang Sci‐Tech University Hangzhou 310018 China
| | - Yanbin Chen
- School of Materials Science and Engineering Zhejiang Sci‐Tech University Hangzhou 310018 China
| | - Jiancang Zhou
- Department of Critical Care Medicine Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou 310016 China
| | - Haomiao Zhang
- Department of Critical Care Medicine Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou 310016 China
- State Key Laboratory of Silicon Materials Key Laboratory of Advanced Materials and Applications for Batteries of Zhejiang Province School of Materials Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Xinhui Xia
- Department of Critical Care Medicine Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou 310016 China
- State Key Laboratory of Silicon Materials Key Laboratory of Advanced Materials and Applications for Batteries of Zhejiang Province School of Materials Science and Engineering Zhejiang University Hangzhou 310027 China
- College of Materials Science and Engineering Zhejiang University of Technology Hangzhou 310014 China
| | - Yefeng Yang
- School of Materials Science and Engineering Zhejiang Sci‐Tech University Hangzhou 310018 China
| | - Yongqi Zhang
- Institute of Fundamental and Frontier Science University of Electronic Science and Technology of China Chengdu 611371 China
| | - Abolhassan Noori
- Department of Chemistry Faculty of Basic Sciences Tarbiat Modares University Tehran 14117‐13116 Iran
| | - Mir F. Mousavi
- Department of Chemistry Faculty of Basic Sciences Tarbiat Modares University Tehran 14117‐13116 Iran
| | - Minghua Chen
- Key Laboratory of Engineering Dielectric and Applications (Ministry of Education) School of Electrical and Electronic Engineering Harbin University of Science and Technology Harbin 150080 P. R. China
| | - Yang Xia
- College of Materials Science and Engineering Zhejiang University of Technology Hangzhou 310014 China
| | - Wenkui Zhang
- College of Materials Science and Engineering Zhejiang University of Technology Hangzhou 310014 China
| |
Collapse
|
89
|
Jia Y, Shi K, Dai L, He X, Deng H, Han R, Yang F, Chu B, Liao J, Wei X, Qian Z. Gold Nanorods and Polymer Micelles Mediated Dual TLR Stimulators Delivery System CPG@Au NRs/M-R848 Regulate Macrophages Reprogramming and DC Maturation for Enhanced Photothermal Immunotherapy of Melanoma. SMALL METHODS 2023; 7:e2201087. [PMID: 36572641 DOI: 10.1002/smtd.202201087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/30/2022] [Indexed: 05/17/2023]
Abstract
Synergistic photothermal immunotherapy has emerged as a favorable therapeutic approach to fight cancer. However, design of an effective photothermal immunotherapy system to suppress tumor growth and simultaneously inhibit tumor metastases continues to be a challenge. Here a dual toll-like receptor agonists delivery system CPG@Au NRs/m-R848 for combined photothermal immunotherapy of melanoma is developed. CPG@Au NRs/m-R848 displays strong antitumor effects by promoting maturation of dendritic cells (DCs) and reprogramming of M2 macrophages into M1 phenotype. Moreover, immunogenic cell death (ICD) induced by photothermal ablation of Au NRs could synergistically produce in situ vaccination effect with CPG ODN and R848, generating systemic and lasting antitumor immunity. It is further proved that CPG@Au NRs/m-R848 treatment inhibits tumor growth in bilateral B16F10 tumors model by eliciting CD8+ T cell response. Overall, this work suggests that this strategy hold great potential in tumor immunotherapy by regulating tumor-associated macrophage polarization, triggering DCs maturation and inducing ICD.
Collapse
Affiliation(s)
- YanPeng Jia
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210000, P. R. China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - LiQun Dai
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - XinLong He
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - HanZhi Deng
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - RuXia Han
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Fan Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - BingYang Chu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - JinFeng Liao
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - XiaWei Wei
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - ZhiYong Qian
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
90
|
Li Y, Yuan R, Luo Y, Guo X, Yang G, Li X, Zhou S. A Hierarchical Structured Fiber Device Remodeling the Acidic Tumor Microenvironment for Enhanced Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300216. [PMID: 36912443 DOI: 10.1002/adma.202300216] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/23/2023] [Indexed: 05/26/2023]
Abstract
The acidic microenvironment of tumors significantly reduces the anti-tumor effect of immunotherapy. Herein, a hierarchically structured fiber device is developed as a local drug delivery system for remodeling the acidic tumor microenvironment (TME) to improve the therapeutic effect of immunotherapy. Proton pump inhibitors in the fiber matrix can be sustainedly released to inhibit the efflux of intracellular H+ from tumor cells, resulting in the remodeling of the acidic TME. The targeted micelles and M1 macrophage membrane-coated nanoparticles in internal cavities of fiber can induce immunogenic cell death (ICD) of tumor cells and phenotypic transformation of tumor-associated macrophages (TAMs), respectively. The relief of the acidity in the TME further promotes ICD and the polarization of TAMs, alleviating the immunosuppressive microenvironment and synergistically enhancing the antitumor immune response. In vivo results reveal this local drug delivery system restores the pH value of TME from 6.8 to 7.2 and exhibit an excellent immunotherapeutic effect.
Collapse
Affiliation(s)
- Yan Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Ruiting Yuan
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yang Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xing Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Guang Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xilin Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
91
|
Perez-Potti A, Rodríguez-Pérez M, Polo E, Pelaz B, Del Pino P. Nanoparticle-based immunotherapeutics: from the properties of nanocores to the differential effects of administration routes. Adv Drug Deliv Rev 2023; 197:114829. [PMID: 37121275 DOI: 10.1016/j.addr.2023.114829] [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: 02/22/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
The engagement with the immune system is one of the main cornerstones in the development of nanotechnologies for therapy and diagnostics. Recent advances have made possible the tuning of features like size, shape and biomolecular modifications that influence such interactions, however, the capabilities for immune modulation of nanoparticles are still not well defined and exploited. This review focuses on recent advances made in preclinical research for the application of nanoparticles to modulate immune responses, and the main features making them relevant for such applications. We review and discuss newest evidence in the field, which include in vivo experiments with an extensive physicochemical characterization as well as detailed study of the induced immune response. We emphasize the need of incorporating knowledge about immune response development and regulation in the design and application of nanoparticles, including the effect by parameters such as the administration route and the differential interactions with immune subsets.
Collapse
Affiliation(s)
- André Perez-Potti
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Manuel Rodríguez-Pérez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
92
|
Li X, Luo Y, Huang Z, Wang Y, Wu J, Zhou S. Multifunctional Liposomes Remodeling Tumor Immune Microenvironment for Tumor Chemoimmunotherapy. SMALL METHODS 2023; 7:e2201327. [PMID: 37075716 DOI: 10.1002/smtd.202201327] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
In the treatment of solid tumors, the complex barriers composed of cancer-associated fibroblasts (CAFs) prevent drug delivery and T cells infiltration into tumor tissues. Although nanocarriers hold great prospects in drug delivery, fibrosis causes the biological barrier and immunosuppressive tumor microenvironment (ITM) that impairs the anti-tumor efficacy of nanocarriers. Here, a small dendritic macromolecule loaded with doxorubicin (PAMAM-ss-DOX) (DP) is synthesized and encapsulated into pH-responsive nanoliposome, together with adjuvant toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) and losartan (LOS). The pH-responsive liposome facilitates the simultaneous and effective delivery of DP, R848, and LOS, which can decompose and release these drugs under the acidic tumor microenvironment. The small sized DP (≈25 nm) with the ability to penetrate into tumor tissue and immunogenic cell death (ICD) can reverse the ITM and elicit immune response, which is equivalent to the effect of an in situ vaccine. Moreover, LOS reduces the activity of CAFs effectively, which can contribute to the infiltration of T cells. Therefore, this nano-platform provides a new therapeutic strategy for enhanced chemo-immunotherapy.
Collapse
Affiliation(s)
- Xinyang Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yang Luo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Zhengjie Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jian Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
93
|
Li D, Zhao A, Zhu J, Wang C, Shen J, Zheng Z, Pan F, Liu Z, Chen Q, Yang Y. Inhaled Lipid Nanoparticles Alleviate Established Pulmonary Fibrosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2300545. [PMID: 37058092 DOI: 10.1002/smll.202300545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/19/2023] [Indexed: 06/19/2023]
Abstract
Pulmonary fibrosis, a sequela of lung injury resulting from severe infection such as severe acute respiratory syndrome-like coronavirus (SARS-CoV-2) infection, is a kind of life-threatening lung disease with limited therapeutic options. Herein, inhalable liposomes encapsulating metformin, a first-line antidiabetic drug that has been reported to effectively reverse pulmonary fibrosis by modulating multiple metabolic pathways, and nintedanib, a well-known antifibrotic drug that has been widely used in the clinic, are developed for pulmonary fibrosis treatment. The composition of liposomes made of neutral, cationic or anionic lipids, and poly(ethylene glycol) (PEG) is optimized by evaluating their retention in the lung after inhalation. Neutral liposomes with suitable PEG shielding are found to be ideal delivery carriers for metformin and nintedanib with significantly prolonged retention in the lung. Moreover, repeated noninvasive aerosol inhalation delivery of metformin and nintedanib loaded liposomes can effectively diminish the development of fibrosis and improve pulmonary function in bleomycin-induced pulmonary fibrosis by promoting myofibroblast deactivation and apoptosis, inhibiting transforming growth factor 1 (TGFβ1) action, suppressing collagen formation, and inducing lipogenic differentiation. Therefore, this work presents a versatile platform with promising clinical translation potential for the noninvasive inhalation delivery of drugs for respiratory disease treatment.
Collapse
Affiliation(s)
- Dongjun Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Ang Zhao
- Department of medical affair, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P. R. China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P. R. China
| | - Jiafei Zhu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Chunjie Wang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Jingjing Shen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Zixuan Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P. R. China
| | - Feng Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P. R. China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P. R. China
| |
Collapse
|
94
|
Liu Y, Niu L, Li N, Wang Y, Liu M, Su X, Bao X, Yin B, Shen S. Bacterial-Mediated Tumor Therapy: Old Treatment in a New Context. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205641. [PMID: 36908053 PMCID: PMC10131876 DOI: 10.1002/advs.202205641] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Targeted therapy and immunotherapy have brought hopes for precision cancer treatment. However, complex physiological barriers and tumor immunosuppression result in poor efficacy, side effects, and resistance to antitumor therapies. Bacteria-mediated antitumor therapy provides new options to address these challenges. Thanks to their special characteristics, bacteria have excellent ability to destroy tumor cells from the inside and induce innate and adaptive antitumor immune responses. Furthermore, bacterial components, including bacterial vesicles, spores, toxins, metabolites, and other active substances, similarly inherit their unique targeting properties and antitumor capabilities. Bacteria and their accessory products can even be reprogrammed to produce and deliver antitumor agents according to clinical needs. This review first discusses the role of different bacteria in the development of tumorigenesis and the latest advances in bacteria-based delivery platforms and the existing obstacles for application. Moreover, the prospect and challenges of clinical transformation of engineered bacteria are also summarized.
Collapse
Affiliation(s)
- Yao Liu
- Key Laboratory of Spine and Spinal Cord Injury Repairand Regeneration of Ministry of EducationOrthopaedic Department of Tongji Hospital, The Institute for Biomedical Engineering and Nano ScienceTongji University School of MedicineShanghai200092P. R. China
- Pharmacy Department and Center for Medical Research and InnovationShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Lili Niu
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Nannan Li
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Yang Wang
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Mingyang Liu
- Department of Surgical Oncology and General SurgeryThe First Hospital of China Medical University155 North Nanjing Street, Heping DistrictShenyang110001China
| | - Xiaomin Su
- Central LaboratoryFirst Affiliated HospitalInstitute (College) of Integrative MedicineDalian Medical UniversityDalian116021China
| | - Xuhui Bao
- Institute for Therapeutic Cancer VaccinesFudan University Pudong Medical CenterShanghai201399China
| | - Bo Yin
- Institute for Therapeutic Cancer Vaccines and Department of OncologyFudan University Pudong Medical CenterShanghai201399China
| | - Shun Shen
- Pharmacy Department and Center for Medical Research and InnovationShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| |
Collapse
|
95
|
Ma J, Guo D, Ji X, Zhou Y, Liu C, Li Q, Zhang J, Fan C, Song H. Composite Hydrogel for Spatiotemporal Lipid Intervention of Tumor Milieu. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211579. [PMID: 36637436 DOI: 10.1002/adma.202211579] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/10/2023] [Indexed: 06/17/2023]
Abstract
Induction of immunogenic cell death (ICD) plays crucial roles in cancer immunotherapy, whereas its efficacy is severely compromised by redundant antioxidant defenses in cancer cells and aberrant lipid metabolism in immunosuppressive cell populations. In this work, it is found that hollow mesoporous CuS nanoparticles (NPs) possess an intrinsic capacity of inhibiting glutathione peroxidase 4 (GPX4). When loaded with an inhibitor of the ferroptosis suppressor protein 1 (FSP1), these NPs block two parallel redox systems and cooperate with near-infrared irradiation to reinforce ICD. A hydrogel co-delivering cancer-cell-targeting CuS NPs and immunosuppressive-cell-targeting sulfo-N-succinimidyl oleate (SSO) for spatiotemporal lipid intervention i further fabricated. While the CuS NPs augment ICD via synergistic lipid peroxidation, SSO reinstates immune perception via lipid metabolic reprogramming, thereby coordinately triggering robust innate and adaptive immunity to restrain tumor growth, relapse, and metastasis. This study provides an immunometabolic therapy via orchestrated lipid modulation in the tumor milieu.
Collapse
Affiliation(s)
- Jia Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Daoxia Guo
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Xiaoyuan Ji
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Yanfeng Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Chang Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| |
Collapse
|
96
|
Ding Y, Qian X, Lin F, Gao B, Wang W, Yang H, Du Y, Wang W. A biomimetic nanoplatform for precise reprogramming of tumor-associated macrophages and NIR-II mediated antitumor immune activation. Acta Biomater 2023; 162:85-97. [PMID: 36948328 DOI: 10.1016/j.actbio.2023.03.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
The therapeutic effects of photothermal therapy (PTT) are dependent on the photothermal conversion efficiency of photothermal agents (PTAs) in tumors and the subsequent activation of the antitumor immune system. However, the insufficient tumor accumulation of current PTAs and the inevitable recruitment of tumor-associated macrophages (TAMs) could further compromise the antitumor activities of PTT. To address these issues, a biomimetic photothermal nanoplatform Au@Fe-PM is developed for the targeted remodeling of TAMs, which promotes the antitumor immunity of PTT. Au nanorods with second near-infrared (NIR-II) absorptions are fabricated to serve as PTAs to induce immunogenic cell death in tumor cells. The ferric hydroxide shell coated on Au nanorods can release iron ions to repolarize M2-like TAMs into the tumoricidal M1 phenotype via P38 and STAT1-mediated signaling pathways. Moreover, the surface decoration of platelet membranes endows biomimetic nanoplatform with enhanced tumor targeting ability for precise tumor ablation and TAM regulation. Consequently, Au@Fe-PM under NIR-II laser irradiation exhibits significantly higher inhibitory effects in a poor immunogenic 4T1 tumor-bearing mouse model with a 50% complete remission rate compared to conventional PTT (0%). By simultaneously reversing the immunosuppressive tumor microenvironment, this biomimetic nanoplatform offers a promising strategy for enhancing the antitumor efficacy of PTT. STATEMENT OF SIGNIFICANCE: The therapeutic effects of current photothermal therapy (PTT) are hindered by the insufficient tumor accumulation of conventional photothermal agents and the recruitment of immunosuppressive tumor-associated macrophages (TAMs) after PTT. Herein, we report a biomimetic iron-based second near-infrared (NIR-II) photothermal nanoplatform (Au@Fe-PM) for targeted TAMs reprogramming and NIR-II mediated anti-tumor immunity. Au@Fe-PM can actively target the tumor site with the help of surface-decorated platelet membranes. Meanwhile, iron ions would be released from Au@Fe-PM in acidic lysosomes to reprogram TAMs into tumoricidal M1-like macrophages, which promotes the antitumor responses elicited by NIR-II PTT, thereby contributing to remarkable tumor inhibitory effects, with 50% higher complete remission rate than that of conventional PTT.
Collapse
Affiliation(s)
- Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang 310058, China
| | - Xiaohui Qian
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang 310058, China
| | - Fenghao Lin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Bingqiang Gao
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang 310058, China
| | - Weili Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang 310058, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China.
| | - Yang Du
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang 310058, China.
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
97
|
Fernandes PD, Magalhães FD, Pereira RF, Pinto AM. Metal-Organic Frameworks Applications in Synergistic Cancer Photo-Immunotherapy. Polymers (Basel) 2023; 15:polym15061490. [PMID: 36987269 PMCID: PMC10053741 DOI: 10.3390/polym15061490] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Conventional cancer therapies, such as radiotherapy and chemotherapy, can have long-term side effects. Phototherapy has significant potential as a non-invasive alternative treatment with excellent selectivity. Nevertheless, its applicability is restricted by the availability of effective photosensitizers and photothermal agents, and its low efficacy when it comes to avoiding metastasis and tumor recurrence. Immunotherapy can promote systemic antitumoral immune responses, acting against metastasis and recurrence; however, it lacks the selectivity displayed by phototherapy, sometimes leading to adverse immune events. The use of metal-organic frameworks (MOFs) in the biomedical field has grown significantly in recent years. Due to their distinct properties, including their porous structure, large surface area, and inherent photo-responsive properties, MOFs can be particularly useful in the fields of cancer phototherapy and immunotherapy. MOF nanoplatforms have successfully demonstrated their ability to address several drawbacks associated with cancer phototherapy and immunotherapy, enabling an effective and low-side-effect combinatorial synergistical treatment for cancer. In the coming years, new advancements in MOFs, particularly regarding the development of highly stable multi-function MOF nanocomposites, may revolutionize the field of oncology.
Collapse
Affiliation(s)
- Pedro D. Fernandes
- LEPABE, Faculdade de Engenharia, Universidade do Porto, Rua Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Fernão D. Magalhães
- LEPABE, Faculdade de Engenharia, Universidade do Porto, Rua Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Artur M. Pinto
- LEPABE, Faculdade de Engenharia, Universidade do Porto, Rua Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
98
|
Yang M, Conceição M, Chen W, Yang F, Zhao B, Wood MJA, Qiu L, Chen J. Engineered bacteria combined with doxorubicin nanoparticles suppress angiogenesis and metastasis in murine melanoma models. Acta Biomater 2023; 158:734-746. [PMID: 36563772 DOI: 10.1016/j.actbio.2022.12.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 12/25/2022]
Abstract
Methods capable of distributing antitumour therapeutics uniformly throughout an entire tumour and that can suppress metastasis at the same time, would be of great significance in improving cancer treatment. Bacteria-mediated synergistic therapies have been explored for better specificity, temporal and spatial controllability, as well for providing regulation of the immune microenvironment, in order to provide improved cancer treatment. To achieve this goal, here we developed an engineered bacteria delivery system (GDOX@HSEc) using synthetic biology and interfacial chemistry technologies. The engineered bacteria were concurrently modified to express heparin sulfatase 1 (HSulf-1) inside (HSEc), to attach doxorubicin-loaded glycogen nanoparticles (GDOX NPs) on their surface. Here we demonstrate that HSEc can actively target and colonise tumour sites resulting in HSulf-1 overexpression, thereby suppressing angiogenesis and metastasis. Simultaneously, the GDOX NPs were able to penetrate into tumour cells, leading to intracellular DNA damage. Our results confirmed that a combination of biotherapy and chemotherapy using GDOX@HSEc resulted in significant melanoma suppression in murine models, with reduced side effects. This study provides a powerful platform for the simultaneous delivery of biomacromolecules and chemotherapeutic drugs to tumours, representing an innovative strategy potentially more effective in treating solid tumours. STATEMENT OF SIGNIFICANCE: An original engineered bacteria-based system (GDOX@HSEc) was developed using synthetic biology and interfacial chemistry technologies to concurrently produce naturally occurring heparin sulfatase 1 (HSulf-1) inside and anchor doxorubicin-loaded glycogen nanoparticles on the surface. GDOX@HSEc allowed for combined local delivery of chemotherapeutic agents along with the enzymes and immunostimulatory bacterial adjuvants, which resulted in a synergistic action in the inhibition of tumour growth and metastasis. The study provides a potential therapeutic approach that allows therapeutic agents to be distributed in a spatiotemporally controllable manner in tumours for combinatorial enhanced therapy.
Collapse
Affiliation(s)
- Meiyang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | | | - Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Fuwei Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Bingke Zhao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, Oxford, UK; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China; Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China.
| |
Collapse
|
99
|
Shen X, Zhu C, Liu X, Zheng H, Wu Q, Xie J, Huang H, Liao Z, Shi J, Nan K, Wang J, Mao X, Gu Z, Li H. Engineered bacteria for augmented in situ tumor vaccination. Biomater Sci 2023; 11:1137-1152. [PMID: 36601796 DOI: 10.1039/d2bm01593e] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In situ tumor vaccination has aroused tremendous interest with its capability for eliciting strong and systemic antitumor immune responses. Unlike traditional cancer vaccines, in situ tumor vaccination avoids the laborious process of tumor antigen identification and can modulate tumor immunosuppressive microenvironment at the same time. In recent years, bacteria have been used as both efficient tumor-targeted delivery vehicles and potent adjuvants. Regarding the rapid development in this area, in this review, we summarize recent advances in the application of bacteria for in situ cancer vaccination. We illustrate the mechanisms of bacteria as both efficient tumor immunogenic cell death inducers and tumor-targeted delivery platforms. Then we comprehensively review the engineering strategies for designing bacteria-based in situ vaccination, including chemical modification, nanotechnology, and genetic engineering. The current dilemma and future directions are discussed at the end of this review.
Collapse
Affiliation(s)
- Xinyuan Shen
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Chaojie Zhu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China. .,Department of Hepatobiliary and Pancreatic Surgery the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Xutao Liu
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Hanqi Zheng
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qing Wu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| | - Jijin Xie
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hao Huang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ziyan Liao
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Jiaqi Shi
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| | - Kewang Nan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Junxia Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Xuming Mao
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China.,Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.,Jinhua Institute of Zhejiang University, Jinhua 321299, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hongjun Li
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China. .,Department of Hepatobiliary and Pancreatic Surgery the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| |
Collapse
|
100
|
Zheng J, Huang J, Zhang L, Wang M, Xu L, Dou X, Leng X, Fang M, Sun Y, Wang Z. Drug-loaded microbubble delivery system to enhance PD-L1 blockade immunotherapy with remodeling immune microenvironment. Biomater Res 2023; 27:9. [PMID: 36759928 PMCID: PMC9909878 DOI: 10.1186/s40824-023-00350-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Although programmed cell death protein 1 (PD-1)/ programmed cell death-ligand protein 1 (PD-L1) checkpoint blockade immunotherapy demonstrates great promise in cancer treatment, poor infiltration of T cells resulted from tumor immunosuppressive microenvironment (TIME) and insufficient accumulation of anti-PD-L1 (αPD-L1) in tumor sites diminish the immune response. Herein, we reported a drug-loaded microbubble delivery system to overcome these obstacles and enhance PD-L1 blockade immunotherapy. METHODS Docetaxel (DTX) and imiquimod (R837)-loaded microbubbles (RD@MBs) were synthesized via a typical rotary evaporation method combined with mechanical oscillation. The targeted release of drugs was achieved by using the directional "bursting" capability of ultrasound-targeted microbubble destruction (UTMD) technology. The antitumor immune response by RD@MBs combining αPD-L1 were evaluated on 4T1 and CT26 tumor models. RESULTS The dying tumor cells induced by DTX release tumor-associated antigens (TAAs), together with R837, promoted the activation, proliferation and recruitment of T cells. Besides, UTMD technology and DTX enhanced the accumulation of αPD-L1 in tumor sites. Moreover, RD@MBs remolded TIME, including the polarization of M2-phenotype tumor-associated macrophages (TAMs) to M1-phenotype, and reduction of myeloid-derived suppressor cells (MDSCs). The RD@MBs + αPD-L1 synergistic therapy not only effectively inhibited the growth of primary tumors, but also significantly inhibited the mimic distant tumors as well as lung metastases. CONCLUSION PD-L1 blockade immunotherapy was enhanced by RD@MBs delivery system.
Collapse
Affiliation(s)
- Jun Zheng
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Ju Huang
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Liang Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China. .,Ultrasound Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People's Republic of China.
| | - Mengna Wang
- grid.203458.80000 0000 8653 0555Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Lihong Xu
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Xiaoyun Dou
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Xiaojing Leng
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Mingxiao Fang
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Yang Sun
- State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| | - Zhigang Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|