1
|
Guan T, Lu Z, Tai R, Guo S, Zhang Z, Deng S, Ye J, Chi K, Zhang B, Chen H, Deng Z, Ke Y, Huang A, Chen P, Wang C, Ou C. Silicified curcumin microspheres Combats cardiovascular diseases via Nrf2/HO-1 pathway. Bioact Mater 2025; 49:378-398. [PMID: 40144796 PMCID: PMC11937612 DOI: 10.1016/j.bioactmat.2025.03.007] [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: 01/17/2025] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Diabetes and chemotherapy frequently give rise to severe cardiovascular complications, including chemotherapy-induced cardiotoxicity and diabetes-associated vascular remodeling. Nevertheless, the precise epidemiological features of these cardiovascular ailments remain incompletely elucidated, resulting in a dearth of effective therapeutic strategies in clinical settings. To tackle this intricate challenge, we have delved extensively into database resources, conducted comprehensive analyses of pertinent epidemiological data, and designed silicified curcumin (Si/Cur) microspheres as a novel therapeutic approach for cardiovascular diseases. By harnessing the alkaline microenvironment generated by silicon (Si), Si/Cur markedly elevates the bioavailability of curcumin (Cur). Further investigations have elucidated that Si/Cur exerts its therapeutic actions primarily via the Nrf2/HO-1 signaling pathway, effectively suppressing vascular remodeling and mitigating myocardial injury, thus disrupting the vicious cycle of persistent cardiovascular damage. In conclusion, this study integrates clinical cohort research to dissect epidemiological characteristics, directs the design and application of biomaterials, and paves the way for a novel and efficacious therapeutic avenue for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Tianwang Guan
- Guangdong Engineering Research Center of Boron Neutron Therapy and Application in Malignant Tumors, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, Dongguan Engineering Research Center for Innovative Boron Drugs and Novel Radioimmune Drugs, Cancer Center, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong, 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
| | - Zhenxing Lu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Rundong Tai
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Shuai Guo
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Zhaowenbin Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shaohui Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Jujian Ye
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Kaiyi Chi
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510182, China
| | - Binghua Zhang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, China
| | - Huiwan Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhilin Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Yushen Ke
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Andong Huang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, China
| | - Peier Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| |
Collapse
|
2
|
Totter E, von Einsiedel E, Regazzoni L, Schuerle S. Paving the way for bacteria-based drug delivery: biohybrid microrobots emerging from microrobotics and synthetic biology. Adv Drug Deliv Rev 2025; 221:115577. [PMID: 40250568 DOI: 10.1016/j.addr.2025.115577] [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: 01/27/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
Advances in microrobotics and synthetic biology are paving the way for innovative solutions to long-standing challenges in drug delivery. Both fields have independently worked on engineering bacteria as a therapeutic system, focusing on enhancing propulsion, cargo delivery, detection, and biocompatibility. Bacteria, with their inherent adaptability and functional versatility, serve as an ideal foundation for these efforts, enabling them to navigate complex biological environments such as the human body. This review explores the convergence of microrobotics and synthetic biology, which has catalysed the development of biohybrid bacterial microrobots that integrate the strengths of both disciplines. By incorporating external control modalities - such as light, ultrasound, and magnetic fields - these hybrid systems address the limitations of purely microrobotic or biological approaches, offering new opportunities to enhance precision and efficacy in targeted therapies. However, realising the full potential of biohybrid bacterial microrobots requires overcoming critical challenges, such as ensuring compatibility between biological and synthetic components, scaling manufacturing processes, and defining regulatory pathways tailored to living therapeutics. Addressing these hurdles through joint, interdisciplinary research efforts, can unlock the transformative possibilities of these systems in modern medicine.
Collapse
Affiliation(s)
- Elena Totter
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland
| | - Emilie von Einsiedel
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland
| | - Lisa Regazzoni
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland
| | - Simone Schuerle
- ETH Zurich, Institute of Translational Medicine, Gloriastrasse 37/39, 8092 Zurich, Switzerland.
| |
Collapse
|
3
|
Kaur T, Devi N, Sharma D. E. coli as a Smart Thermo-Vector for Combating Solid Tumors: A Synergistic Heat-Induced Cancer Therapy Approach. Bioconjug Chem 2025; 36:867-880. [PMID: 40105793 DOI: 10.1021/acs.bioconjchem.5c00102] [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: 03/20/2025]
Abstract
Heat-induced cancer therapies such as magnetic hyperthermia-based cancer therapy (MHCT) and photothermal tumor ablation (PTT) have garnered significant attention as minimally invasive new-generation cancer therapy modalities. However, solid tumors associated with hypoxia present a considerable challenge to effective cancer therapy. In this study, we took up the challenge of mitigating the limiting penetration ability of nanoparticles by integrating polydopamine-coated magnetic nanoparticles and motile anaerobic bacteria (PDBs) to function as a smart thermo-vector. The developed PDBs are capable of self-navigating hypoxic tumors and as thermo-therapy agents with the ability to induce heat through exposure to an alternating magnetic field or near-infrared laser light. The thermo-vector system exhibited a dual-functioning synergistic antitumor effect of MHCT and PTT and an outstanding tumor targeting efficiency, outperforming the conventional 'nanoparticles only' approach. The heat-induced cellular oxidative stress and disrupted mitochondrial function led to 80% cellular cytotoxicity within 24 h of treatment. The PDB-based approach led to complete tumor regression in c57BL/6 mice within 21 days of treatment and a tumor-free survival for 60 days without recurrence, proving the capability of the developed PDBs in combatting solid tumors.
Collapse
Affiliation(s)
- Tashmeen Kaur
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Neeta Devi
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Deepika Sharma
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| |
Collapse
|
4
|
Ran H, Yang Y, Han W, Liang R, Zhu D, Yuan B, Xu C, Li D, Ren J, Pan H, Liu L, Ma T, Ma A, Cai L. Programmable ultrasound-mediated swarms manipulation of bacteria-red blood cell microrobots for tumor-specific thrombosis and robust photothermal therapy. Trends Biotechnol 2025; 43:868-892. [PMID: 39709244 DOI: 10.1016/j.tibtech.2024.11.018] [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: 03/31/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
Despite the excellent advantages of biomicrorobots, such as autonomous navigation and targeting actuation, effective penetration and retention to deep lesion sites for effective therapy remains a longstanding challenge. Here, we present dual-engine cell microrobots, which we refer to as PR-robots, created by conjugating photosynthetic bacteria (PSB) with red blood cells (RBCs). The robots penetrate the tumor interior in swarms through combined hypoxic traction and ultrasound actuation (UA). The hypoxia-targeting ability of PSB induced PR-robot accumulation in the tumor region. Subsequently, programmable UA trapped the PR-robots to form bioswarms and traverse tissue obstacles, penetrating the tumor interior. The substantial influx of PR-robots into the tumor tissue promoted the formation of tumor-specific thrombus (TST). Finally, the PSB and TST synergistically improved the effect of photothermal therapy. Thus, these advantages of remote ultrasound control technology pave the way for various new therapies in practical biomedicine.
Collapse
Affiliation(s)
- Hui Ran
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan 523808, PR China
| | - Ye Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Weijing Han
- Songshan Lake Materials Laboratory, Dongguan 523808, PR China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Denghui Zhu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, PR China
| | - Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan 523808, PR China
| | - Dan Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan 523808, PR China
| | - Jian Ren
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Teng Ma
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Aiqing Ma
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan 523808, PR China.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; Sino-Euro Center of Biomedicine and Health, Shenzhen 518024, PR China.
| |
Collapse
|
5
|
Deng S, Hu L, Chen G, Ye J, Xiao Z, Guan T, Guo S, Xia W, Cheng D, Wan X, Cheng K, Ou C. A PD-L1 siRNA-Loaded Boron Nanoparticle for Targeted Cancer Radiotherapy and Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419418. [PMID: 39955653 DOI: 10.1002/adma.202419418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/25/2025] [Indexed: 02/17/2025]
Abstract
Although the combination of radiotherapy and immunotherapy is regarded as a promising clinical treatment strategy, numerous clinical trials have failed to demonstrate synergistic effects. One of the key reasons is that conventional radiotherapies inevitably damage intratumoral effector immune cells. Boron Neutron Capture Therapy (BNCT) is a precise radiotherapy that selectively kills tumor cells while sparing adjacent normal cells, by utilizing 10B agents and neutron irradiation. Therefore, combinational BNCT-immunotherapy holds promise for achieving more effective synergistic effects. Here it develops a 10B-containing polymer that self-assembled with PD-L1 siRNA to form 10B/siPD-L1 nanoparticles for combinational BNCT-immunotherapy. Unlike antibodies, PD-L1 siRNA can inhibit intracellular PD-L1 upregulated by BNCT, activating T-cell immunity while also suppressing DNA repair. This can enhance BNCT-induced DNA damage, promoting immunogenic cell death (ICD) and further amplifying the antitumor immune effect. The results demonstrated that BNCT using 10B/siPD-L1 nanoparticles precisely killed tumor cells while sparing adjacent T cells and induced a potent antitumor immune response, inhibiting distal and metastatic tumors.
Collapse
Affiliation(s)
- Shaohui Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Lijun Hu
- The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Guo Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jujian Ye
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zecong Xiao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tianwang Guan
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Shuai Guo
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Wei Xia
- Neuboron Medtech Ltd, Nanjing, 211112, China
| | - Du Cheng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, 10032, USA
| | - Caiwen Ou
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| |
Collapse
|
6
|
Zeng SM, Qu WQ, Sun YL, Chen KW, Zhao K, Yan JH, Zhang C, Liang CX, Chen Y, Pan T, Yu A, Zhang XZ. MnO 2-Assisted Photosynthetic Bacteria Interfering with the Adenosine-A2AR Metabolic Pathway to Enhance Tumor Photothermal Immunotherapy. ACS NANO 2025; 19:7962-7980. [PMID: 39976374 DOI: 10.1021/acsnano.4c15139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Hypoxia-related adenosine (Ado) exerts an immunosuppressive effect in tumors by binding to the metabolic checkpoint Ado A2A receptors (A2AR), thereby hindering the activation of antitumor immunity induced by immunogenic cell death (ICD). In this study, a MnO2-assisted photosynthetic bacteria (PSB) biohybrid (MnO2@PSB) is developed to enhance tumor photothermal immunotherapy by interfering with the Ado-A2AR metabolic pathway. Specifically, manganese dioxide (MnO2) nanoflowers are conjugated onto PSB by the carbodiimide reaction to construct the biohybrid MnO2@PSB. As a photothermal agent, MnO2@PSB generates heat to "burn" tumor cells under 808 nm laser irradiation, inducing tumor cell ICD. Meanwhile, MnO2@PSB catalyzes the decomposition of endogenous hydrogen peroxide into oxygen to alleviate tumor hypoxia, thereby reducing Ado production and downregulating the expression of A2AR, further reversing the tumor immunosuppressive microenvironment and amplifying the ICD effects. In various mouse 4T1 tumor models, MnO2@PSB can enhance antitumor immune responses, prolong mouse survival, and significantly inhibit tumor growth, recurrence, and metastasis under 808 nm laser irradiation. Collectively, this study provides a direction for enhanced antitumor immunotherapy through regulating metabolic pathways.
Collapse
Affiliation(s)
- Si-Min Zeng
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Wen-Qiang Qu
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu-Liang Sun
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Ke-Wei Chen
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Kai Zhao
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Jian-Hua Yan
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Cheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Chun-Xiao Liang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu Chen
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Ting Pan
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
7
|
Zhu D, Pan W, Li H, Hua J, Zhang C, Zhao K. Innovative Applications of Bacteria and Their Derivatives in Targeted Tumor Therapy. ACS NANO 2025; 19:5077-5109. [PMID: 39874477 DOI: 10.1021/acsnano.4c15237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Despite significant progress in cancer treatment, traditional therapies still face considerable challenges, including poor targeting, severe toxic side effects, and the development of resistance. Recent advances in biotechnology have revealed the potential of bacteria and their derivatives as drug delivery systems for tumor therapy by leveraging their biological properties. Engineered bacteria, including Escherichia coli, Salmonella, and Listeria monocytogenes, along with their derivatives─outer membrane vesicles (OMVs), bacterial ghosts (BGs), and bacterial spores (BSPs)─can be loaded with a variety of antitumor agents, enabling precise targeting and sustained drug release within the tumor microenvironment (TME). These bacteria and their derivatives possess intrinsic properties that stimulate the immune system, enhancing both innate and adaptive immune responses to further amplify therapeutic effects. The ability of bacteria to naturally accumulate in hypoxic tumor regions and their versatility in genetic modifications allow for tailored drug delivery strategies that synergistically enhance the effectiveness of chemotherapy, immunotherapy, and targeted therapies. This review comprehensively examines the fundamental principles of bacterial therapy, focusing on the strategies employed for bacterial engineering, drug loading, and the use of bacteria and their derivatives in targeted tumor therapy. It also discusses the challenges faced in optimizing bacterial delivery systems, such as safety concerns, unintended immune responses, and scalability for clinical applications. By exploring these aspects, this review provides a theoretical framework for improving bacterial-based drug delivery systems, contributing to the development of more effective and personalized cancer treatments.
Collapse
Affiliation(s)
- Denghui Zhu
- Institute of Nanobiomaterials and Immunology & Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, School of Life Sciences, Taizhou University, Zhejiang Taizhou 318000, China
| | - Wendi Pan
- Institute of Nanobiomaterials and Immunology & Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, School of Life Sciences, Taizhou University, Zhejiang Taizhou 318000, China
| | - Heqi Li
- Institute of Nanobiomaterials and Immunology & Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, School of Life Sciences, Taizhou University, Zhejiang Taizhou 318000, China
- School of Medical Technology, Qiqihar Medical University, Heilongjiang Qiqihar 161006, China
| | - Jingsheng Hua
- Department of Hematology, Municipal Hospital Affiliated to Taizhou University, Zhejiang Taizhou 318000, China
| | - Chunjing Zhang
- School of Medical Technology, Qiqihar Medical University, Heilongjiang Qiqihar 161006, China
| | - Kai Zhao
- Institute of Nanobiomaterials and Immunology & Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, School of Life Sciences, Taizhou University, Zhejiang Taizhou 318000, China
- Department of Hematology, Municipal Hospital Affiliated to Taizhou University, Zhejiang Taizhou 318000, China
| |
Collapse
|
8
|
Cao Y, Zheng M, Shi J, Si J, Huang G, Ji Y, Hou Y, Ge Z. X-ray-Triggered Activation of Polyprodrugs for Synergistic Radiochemotherapy. Biomacromolecules 2025; 26:579-590. [PMID: 39727263 DOI: 10.1021/acs.biomac.4c01373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
X-ray-induced photodynamic therapy (XPDT) can penetrate deeply into the tumor tissues to overcome the disadvantage of conventional PDT. However, the therapeutic efficacy of XPDT in cancer therapy is still restricted due to the insufficient reactive oxygen species (ROS) generation at a relatively low irradiation dosage. Herein, we present the tumor pH and ROS-responsive polyprodrug micelles to load the X-ray photosensitizer verteporfin (VP) as an ROS production enhancer. The block copolymer polyprodrug consisting of hydrophilic poly(ethylene glycol) (PEG) as well as the segments of thioketal-linked camptothecin (CPT) methacrylate (CPTKMA) and 2-(pentamethyleneimino)ethyl methacrylate (PEMA) (PEG-b-P(CPTKMA-co-PEMA)) can self-assemble into micelles in aqueous solution and encapsulate VP with a high loading efficiency of 67%. Inside tumor tissues, the zeta potential of the micelles can transform from neutral to positive for promoted cellular internalization under tumor acidity. Followed by X-ray irradiation at the dose of 4 Gy, efficient ROS generation in the presence of VP triggers CPT release. The VP-loaded polyprodrug micelles can finally ablate tumors efficiently via synergistic radiochemotherapy due to deep penetration of X-ray inside tumor tissues, ROS generation enhancement, and triggered CPT release. Consequently, this promising strategy represents a robust therapeutic modality for the enhanced radiochemotherapy of cancers.
Collapse
Affiliation(s)
- Yufei Cao
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Moujiang Zheng
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Jiahong Shi
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jiale Si
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Guopu Huang
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yuanyuan Ji
- Department of Geriatric General Surgery, Scientific Research Center and Precision Medical Institute, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
9
|
Jiang Y. Photosynthetic Bacteria: Light-Responsive Biomaterials for Anti-Tumor Photodynamic Therapy. Int J Nanomedicine 2025; 20:465-482. [PMID: 39811429 PMCID: PMC11730521 DOI: 10.2147/ijn.s500314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
Photodynamic therapy (PDT) is a promising noninvasive tumor treatment modality that relies on generating reactive oxygen species (ROS) and requires an adequate oxygen supply to the target tissue. However, hypoxia is a common feature of solid tumors and profoundly restricts the anti-tumor efficacy of PDT. In recent years, scholars have focused on exploring nanomaterial-based strategies for oxygen supplementation and integrating non-oxygen-consuming treatment approaches to overcome the hypoxic limitations of PDT. Some scholars have harnessed the photosynthetic oxygen production of cyanobacteria under light irradiation to overcome tumor hypoxia and engineered them as carriers of photosensitizers instead of inorganic nanomaterials, resulting in photosynthetic bacteria (PSB) attracting significant attention. Recent studies have shown that light-triggered PSB can exhibit additional properties, such as photosynthetic hydrogen production, ROS generation, and photothermal conversion, facilitating their use as promising light-responsive biomaterials for enhancing the anti-tumor efficacy of PDT. Therefore, understanding PSB can provide new insights and ideas for future research. This review mainly introduces the characteristics of PSB and recent research on light-triggered PSB in anti-tumor PDT to enrich our knowledge in this area. Finally, the challenges and prospects of using PSB to enhance the anti-tumor efficacy of PDT were also discussed.
Collapse
Affiliation(s)
- Yuan Jiang
- Department of Rehabilitation Medicine, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
10
|
Wang H, Hao D, Wu Q, Sun T, Xie Z. A morphologically transformable hypoxia-induced radical anion for tumor-specific photothermal therapy. Acta Pharm Sin B 2024; 14:5407-5417. [PMID: 39807323 PMCID: PMC11725169 DOI: 10.1016/j.apsb.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 01/16/2025] Open
Abstract
Tumor microenvironment activatable therapeutic agents and their effective tumor accumulation are significant for selective tumor treatment. Herein, we provide an unadulterated nanomaterial combining the above advantages. We synthesize a perylene diimide (PDI) molecule substituted by glutamic acid (Glu), which can self-assemble into small spherical nanoparticles (PDI-SG) in aqueous solution. PDI-SG can not only be transformed into nanofibers at low pH conditions but also be reduced to PDI radical anion (PDI·‒), which exhibits strong near-infrared absorption and excellent photothermal performance. More importantly, PDI-SG can also be reduced to PDI·‒ in hypoxic tumors to ablate the tumors and minimize the damage to normal tissues. The morphological transformation from small nanoparticles to nanofibers makes for better tumor accumulation and retention. This work sheds light on the design of tumor microenvironment activatable therapeutics with precise structures for high-performance tumor therapy.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Dengyuan Hao
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Qihang Wu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Tingting Sun
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
11
|
Li Y, Si Y, Yin H. Nanomaterial-mediated photothermal therapy modulates tumor-associated macrophages: applications in cancer therapy. J Mater Chem B 2024; 12:11867-11886. [PMID: 39501854 DOI: 10.1039/d4tb01928h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Complex pathogenesis and diverse clinical features pose many challenges in selecting appropriate cancer treatment strategies. Recent studies have shown that tumor-associated macrophages (TAMs) play dual roles in both promoting and inhibiting tumor growth. TAMs not only contribute to tumor survival and metastasis but also impact the response to therapy. Nanomaterial-based photothermal therapy (PTT) strategies have been widely used as ablative therapies for various cancers. Many studies have demonstrated that nanomaterial-mediated PTT effectively shifts TAMs towards an anticancer phenotype, thus inducing tumor apoptosis. Therefore, a comprehensive understanding of the tumor immune microenvironment will undoubtedly accelerate advancements in tumor therapy. This paper summarizes the application of nanomaterial-mediated PTT for cancer treatment by modulating TAMs. It highlights the types of nanomaterials and near-infrared laser modes used in the treatment process, analyzes the physicochemical factors that influence the distribution of different isoforms in TAMs, and finally explores the specific therapeutic parameters and mechanisms of nanomaterial-mediated PTT to guide future research in related fields.
Collapse
Affiliation(s)
- Yan Li
- School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Yuhao Si
- School of Acupuncture-Moxibustion and Tuina, School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Heng Yin
- Department of Traumatology & Orthopedics, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi 214071, China.
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu Province 214071, China
| |
Collapse
|
12
|
Sun M, Wang T, Zhu Y, Ling F, Bai J, Tang C. Gas immnuo-nanomedicines fight cancers. Biomed Pharmacother 2024; 180:117595. [PMID: 39476762 DOI: 10.1016/j.biopha.2024.117595] [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: 07/23/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/14/2024] Open
Abstract
Certain gas molecules, including hydrogen (H2), nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), oxygen (O2) and sulfur dioxide (SO2) exhibit significant biological functionalities that can modulate the immune response. Strategies pertaining to gas-based immune therapy have garnered considerable attention in recent years. Nevertheless, delivering various gas molecules precisely into tumors, which leads to enhanced anti-tumor immunotherapeutic effect, is still a main challenge. The advent of gas treatment modality with desirable immunotherapeutic efficiency has been made possible by the rapid development of nanotechnology, which even derives the concept of the gas immnuo-nanomedicines (GINMs). In light of the fact, we herein aim to furnish a cutting-edge review on the latest progress of GINMs. The underlying mechanisms of action for several gases utilized in cancer immunotherapy are initially outlined. Additionally, it provides a succinct overview of the current clinical landscape of gas therapy, and introduces GINMs specifically designed for cancer treatment based on immunotherapeutic principles across multiple strategies. Last but not least, we address the challenges and opportunities associated with GINMs, exploring the potential future developments and clinical applications of this innovative approach.
Collapse
Affiliation(s)
- Mengchi Sun
- Huzhou Key Laboratory of Translational Medicine, Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; College of Art and Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Tianye Wang
- Department of General Surgery, The First Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yinmei Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Feng Ling
- Huzhou Key Laboratory of Translational Medicine, Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Jingwen Bai
- College of Art and Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Chengwu Tang
- Huzhou Key Laboratory of Translational Medicine, Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China.
| |
Collapse
|
13
|
Wang C, Feng Q, Shi S, Qin Y, Lu H, Zhang P, Liu J, Chen B. The Rational Engineered Bacteria Based Biohybrid Living System for Tumor Therapy. Adv Healthc Mater 2024; 13:e2401538. [PMID: 39051784 DOI: 10.1002/adhm.202401538] [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: 04/26/2024] [Revised: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Living therapy based on bacterial cells has gained increasing attention for their applications in tumor treatments. Bacterial cells can naturally target to tumor sites and active the innate immunological responses. The intrinsic advantages of bacteria attribute to the development of biohybrid living carriers for targeting delivery toward hypoxic environments. The rationally engineered bacterial cells integrate various functions to enhance the tumor therapy and reduce toxic side effects. In this review, the antitumor effects of bacteria and their application are discussed as living therapeutic agents across multiple antitumor platforms. The various kinds of bacteria used for cancer therapy are first introduced and demonstrated the mechanism of antitumor effects as well as the immunological effects. Additionally, this study focused on the genetically modified bacteria for the production of antitumor agents as living delivery system to treat cancer. The combination of living bacterial cells with functional nanomaterials is then discussed in the cancer treatments. In brief, the rational design of living therapy based on bacterial cells highlighted a rapid development in tumor therapy and pointed out the potentials in clinical applications.
Collapse
Affiliation(s)
- Chen Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Qiliner Feng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Si Shi
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Yuxuan Qin
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Hongli Lu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Peng Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Baizhu Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
14
|
Chuang AEY, Tao YK, Dong SW, Nguyen HT, Liu CH. Polypyrrole/iron-glycol chitosan nanozymes mediate M1 macrophages to enhance the X-ray-triggered photodynamic therapy for bladder cancer by promoting antitumor immunity. Int J Biol Macromol 2024; 280:135608. [PMID: 39276877 DOI: 10.1016/j.ijbiomac.2024.135608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
X-ray Photodynamic Therapy (XPDT) is an emerging, deeply penetrating, and non-invasive tumor treatment that stimulates robust antitumor immune responses. However, its efficacy is often limited by low therapeutic delivery and immunosuppressant within the tumor microenvironment. This challenge can potentially be addressed by utilizing X-ray responsive iron-glycol chitosan-polypyrrole nanozymes (GCS-I-PPy NZs), which activate M1 macrophages. These nanozymes increase tumor infiltration and enhance the macrophages' intrinsic immune response and their ability to stimulate adaptive immunity. Authors have designed biocompatible, photosensitizer-containing GCS-I-PPy NZs using oxidation/reduction reactions. These nanozymes were internalized by M1 macrophages to form RAW-GCS-I-PPy NZs. Authors' results demonstrated that these engineered macrophages effectively delivered the nanozymes with potentially high tumor accumulation. Within the tumor microenvironment, the accumulated GCS-I-PPy NZs underwent X-ray irradiation, generating reactive oxygen species (ROS). This ROS augmentation significantly enhanced the therapeutic effect of XPDT and synergistically promoted T cell infiltration into the tumor. These findings suggest that nano-engineered M1 macrophages can effectively boost the immune effects of XPDT, providing a promising strategy for enhancing cancer immunotherapy. The ability of GCS-I-PPy NZs to mediate M1 macrophage activation and increase tumor infiltration highlights their potential in overcoming the limitations of current XPDT approaches and improving therapeutic outcomes in melanoma and other cancers.
Collapse
Affiliation(s)
- Andrew E-Y Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan; Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan.
| | - Yu-Kuang Tao
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
| | - Shao-Wei Dong
- Taipei Medical University Shuang Ho Hospital, Taiwan
| | - Hieu Trung Nguyen
- Department of Orthopedics and Trauma, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Viet Nam
| | - Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan; Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
| |
Collapse
|
15
|
Qi Z, Gu J, Qu L, Shi X, He Z, Sun J, Tan L, Sun M. Advancements of engineered live oncolytic biotherapeutics (microbe/virus/cells): Preclinical research and clinical progress. J Control Release 2024; 375:209-235. [PMID: 39244159 DOI: 10.1016/j.jconrel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The proven efficacy of immunotherapy in fighting tumors has been firmly established, heralding a new era in harnessing both the innate and adaptive immune systems for cancer treatment. Despite its promise, challenges such as inefficient delivery, insufficient tumor penetration, and considerable potential toxicity of immunomodulatory agents have impeded the advancement of immunotherapies. Recent endeavors in the realm of tumor prophylaxis and management have highlighted the use of living biological entities, including bacteria, oncolytic viruses, and immune cells, as a vanguard for an innovative class of live biotherapeutic products (LBPs). These LBPs are gaining recognition for their inherent ability to target tumors. However, these LBPs must contend with significant barriers, including robust immune clearance mechanisms, cytotoxicity and other in vivo adverse effects. Priority must be placed on enhancing their safety and therapeutic indices. This review consolidates the latest preclinical research and clinical progress pertaining to the exploitation of engineered biologics, spanning bacteria, oncolytic viruses, immune cells, and summarizes their integration with combination therapies aimed at circumventing current clinical impasses. Additionally, the prospective utilities and inherent challenges of the biotherapeutics are deliberated, with the objective of accelerating their clinical application in the foreseeable future.
Collapse
Affiliation(s)
- Zhengzhuo Qi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Junmou Gu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihang Qu
- The 4th People's Hospital of Shenyang, China Medical University, Shenyang, Liaoning, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Lingchen Tan
- School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
16
|
Lu J, Tong Q. From pathogenesis to treatment: the impact of bacteria on cancer. Front Microbiol 2024; 15:1462749. [PMID: 39360320 PMCID: PMC11445166 DOI: 10.3389/fmicb.2024.1462749] [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: 07/10/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
The intricate relationship between cancer and bacteria has garnered increasing attention in recent years. While traditional cancer research has primarily focused on tumor cells and genetic mutations, emerging evidence highlights the significant role of microbial communities within the tumor microenvironment in cancer development and progression. This review aims to provide a comprehensive overview of the current understanding of the complex interplay between cancer and bacteria. We explore the diverse ways in which bacteria influence tumorigenesis and tumor behavior, discussing direct interactions between bacteria and tumor cells, their impact on tumor immunity, and the potential modulation of the tumor microenvironment. Additionally, we delve into the mechanisms through which bacterial metabolites and extracellular products May affect cancer pathways. By conducting a thorough analysis of the existing literature, we underscore the multifaceted and intricate relationship between bacteria and cancer. Understanding this complex interplay could pave the way for novel therapeutic approaches and preventive strategies in cancer treatment.
Collapse
Affiliation(s)
| | - Qiang Tong
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Ma Y, Hu Y, Liu H, Li X, Li Y, Zhao Y, Zhang Q, Zhang Z, Leng Q, Luo L, Li L, Dai Y, Chen G, Zhang J, Li Z. High-Lactate-Metabolizing Photosynthetic Bacteria Reprogram Tumor Immune Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405930. [PMID: 38924191 DOI: 10.1002/adma.202405930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Indexed: 06/28/2024]
Abstract
The elevated levels of lactate in tumor tissue play a pivotal role in fostering an immunosuppressive microenvironment. Therefore, efficiently reducing lactate levels to reprogram tumor immune microenvironment (TIM) is considered a crucial step for boosted immunotherapy. Here, a high-lactate-metabolizing photosynthetic bacteria (LAB-1) is selectively screened for TIM reprogramming, which then improves the efficacy of tumor immunotherapy. The culture medium for LAB-1 screening is initially developed through an orthogonal experiment, simulating the tumor microenvironment (TME) and utilizing lactate as the sole organic carbon source. As demonstrated in a murine 4T1 model, LAB-1 colonizes the TME selectively, resulting in a significant reduction in lactate levels and a subsequent increase in pH values within the tumor tissue. Furthermore, single-cell RNA sequencing analysis reveals that LAB-1 effectively reprograms the TIM, thereby enhancing the effectiveness of antitumor immune therapy. This approach of utilizing lactate-consuming bacteria represents a potent tool for augmenting tumor immunotherapy efficiency.
Collapse
Affiliation(s)
- Yichuan Ma
- College of Chemistry & Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding, 071002, China
| | - Yujing Hu
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Huifang Liu
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Xiaoya Li
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Yuanhang Li
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Yu Zhao
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Qi Zhang
- College of Chemistry & Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding, 071002, China
| | - Ziyang Zhang
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Qingqing Leng
- College of Pharmaceutical Science, Hebei University, Baoding, 071002, China
| | - Li Luo
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| | - Lanya Li
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, 999078, China
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Jinchao Zhang
- College of Chemistry & Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding, 071002, China
| | - Zhenhua Li
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong, 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| |
Collapse
|
18
|
Liu J, Li B, Li L, Ming X, Xu ZP. Advances in Nanomaterials for Immunotherapeutic Improvement of Cancer Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403024. [PMID: 38773882 DOI: 10.1002/smll.202403024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/10/2024] [Indexed: 05/24/2024]
Abstract
Immuno-stimulative effect of chemotherapy (ISECT) is recognized as a potential alternative to conventional immunotherapies, however, the clinical application is constrained by its inefficiency. Metronomic chemotherapy, though designed to overcome these limitations, offers inconsistent results, with effectiveness varying based on cancer types, stages, and patient-specific factors. In parallel, a wealth of preclinical nanomaterials holds considerable promise for ISECT improvement by modulating the cancer-immunity cycle. In the area of biomedical nanomaterials, current literature reviews mainly concentrate on a specific category of nanomaterials and nanotechnological perspectives, while two essential issues are still lacking, i.e., a comprehensive analysis addressing the causes for ISECT inefficiency and a thorough summary elaborating the nanomaterials for ISECT improvement. This review thus aims to fill these gaps and catalyze further development in this field. For the first time, this review comprehensively discusses the causes of ISECT inefficiency. It then meticulously categorizes six types of nanomaterials for improving ISECT. Subsequently, practical strategies are further proposed for addressing inefficient ISECT, along with a detailed discussion on exemplary nanomedicines. Finally, this review provides insights into the challenges and perspectives for improving chemo-immunotherapy by innovations in nanomaterials.
Collapse
Affiliation(s)
- Jie Liu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 000000, China
- GoodMedX Tech Limited Company, Hong Kong SAR, 000000, China
| | - Bei Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xin Ming
- Departments of Cancer Biology and Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27157, USA
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
- Institute of Biomedical Health Technology and Engineering, and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, 518107, China
| |
Collapse
|
19
|
Liu J, He C, Tan W, Zheng JH. Path to bacteriotherapy: From bacterial engineering to therapeutic perspectives. Life Sci 2024; 352:122897. [PMID: 38971366 DOI: 10.1016/j.lfs.2024.122897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The major reason for the failure of conventional therapies is the heterogeneity and complexity of tumor microenvironments (TMEs). Many malignant tumors reprogram their surface antigens to evade the immune surveillance, leading to reduced antigen-presenting cells and hindered T-cell activation. Bacteria-mediated cancer immunotherapy has been extensively investigated in recent years. Scientists have ingeniously modified bacteria using synthetic biology and nanotechnology to enhance their biosafety with high tumor specificity, resulting in robust anticancer immune responses. To enhance the antitumor efficacy, therapeutic proteins, cytokines, nanoparticles, and chemotherapeutic drugs have been efficiently delivered using engineered bacteria. This review provides a comprehensive understanding of oncolytic bacterial therapies, covering bacterial design and the intricate interactions within TMEs. Additionally, it offers an in-depth comparison of the current techniques used for bacterial modification, both internally and externally, to maximize their therapeutic effectiveness. Finally, we outlined the challenges and opportunities ahead in the clinical application of oncolytic bacterial therapies.
Collapse
Affiliation(s)
- Jinling Liu
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha 410082, China; College of Biology, Hunan University, Changsha 410082, China
| | - Chongsheng He
- College of Biology, Hunan University, Changsha 410082, China
| | - Wenzhi Tan
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha, Hunan 410114, China.
| | - Jin Hai Zheng
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha 410082, China.
| |
Collapse
|
20
|
Peng F, Hu M, Su Z, Hu L, Guo L, Yang K. Intratumoral Microbiota as a Target for Advanced Cancer Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405331. [PMID: 39054925 DOI: 10.1002/adma.202405331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/21/2024] [Indexed: 07/27/2024]
Abstract
In recent years, advancements in microbial sequencing technology have sparked an increasing interest in the bacteria residing within solid tumors and its distribution and functions in various tumors. Intratumoral bacteria critically modulate tumor oncogenesis and development through DNA damage induction, chronic inflammation, epigenetic alterations, and metabolic and immune regulation, while also influencing cancer treatment efficacy by affecting drug metabolism. In response to these discoveries, a variety of anti-cancer therapies targeting these microorganisms have emerged. These approaches encompass oncolytic therapy utilizing tumor-associated bacteria, the design of biomaterials based on intratumoral bacteria, the use of intratumoral bacterial components for drug delivery systems, and comprehensive strategies aimed at the eradication of tumor-promoting bacteria. Herein, this review article summarizes the distribution patterns of bacteria in different solid tumors, examines their impact on tumors, and evaluates current therapeutic strategies centered on tumor-associated bacteria. Furthermore, the challenges and prospects for developing drugs that target these bacterial communities are also explored, promising new directions for cancer treatment.
Collapse
Affiliation(s)
- Fei Peng
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Mengyuan Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhiyue Su
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lingchuan Guo
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Kai Yang
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Alkene-carbon Fibres-based Technology & Application for Detection of Major Infectious Diseases, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
21
|
Chen M, Xia L, Wu C, Wang Z, Ding L, Xie Y, Feng W, Chen Y. Microbe-material hybrids for therapeutic applications. Chem Soc Rev 2024; 53:8306-8378. [PMID: 39005165 DOI: 10.1039/d3cs00655g] [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: 07/16/2024]
Abstract
As natural living substances, microorganisms have emerged as useful resources in medicine for creating microbe-material hybrids ranging from nano to macro dimensions. The engineering of microbe-involved nanomedicine capitalizes on the distinctive physiological attributes of microbes, particularly their intrinsic "living" properties such as hypoxia tendency and oxygen production capabilities. Exploiting these remarkable characteristics in combination with other functional materials or molecules enables synergistic enhancements that hold tremendous promise for improved drug delivery, site-specific therapy, and enhanced monitoring of treatment outcomes, presenting substantial opportunities for amplifying the efficacy of disease treatments. This comprehensive review outlines the microorganisms and microbial derivatives used in biomedicine and their specific advantages for therapeutic application. In addition, we delineate the fundamental strategies and mechanisms employed for constructing microbe-material hybrids. The diverse biomedical applications of the constructed microbe-material hybrids, encompassing bioimaging, anti-tumor, anti-bacteria, anti-inflammation and other diseases therapy are exhaustively illustrated. We also discuss the current challenges and prospects associated with the clinical translation of microbe-material hybrid platforms. Therefore, the unique versatility and potential exhibited by microbe-material hybrids position them as promising candidates for the development of next-generation nanomedicine and biomaterials with unique theranostic properties and functionalities.
Collapse
Affiliation(s)
- Meng Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Li Ding
- Department of Medical Ultrasound, National Clinical Research Center of Interventional Medicine, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Tongji University, Shanghai, 200072, P. R. China.
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| |
Collapse
|
22
|
Gao P, Duan Z, Xu G, Gong Q, Wang J, Luo K, Chen J. Harnessing and Mimicking Bacterial Features to Combat Cancer: From Living Entities to Artificial Mimicking Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405075. [PMID: 39136067 DOI: 10.1002/adma.202405075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Bacterial-derived micro-/nanomedicine has garnered considerable attention in anticancer therapy, owing to the unique natural features of bacteria, including specific targeting ability, immunogenic benefits, physicochemical modifiability, and biotechnological editability. Besides, bacterial components have also been explored as promising drug delivery vehicles. Harnessing these bacterial features, cutting-edge physicochemical and biotechnologies have been applied to attenuated tumor-targeting bacteria with unique properties or functions for potent and effective cancer treatment, including strategies of gene-editing and genetic circuits. Further, the advent of bacteria-inspired micro-/nanorobots and mimicking artificial systems has furnished fresh perspectives for formulating strategies for developing highly efficient drug delivery systems. Focusing on the unique natural features and advantages of bacteria, this review delves into advances in bacteria-derived drug delivery systems for anticancer treatment in recent years, which has experienced a process from living entities to artificial mimicking systems. Meanwhile, a summary of relative clinical trials is provided and primary challenges impeding their clinical application are discussed. Furthermore, future directions are suggested for bacteria-derived systems to combat cancer.
Collapse
Affiliation(s)
- Peng Gao
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Gang Xu
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kui Luo
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Jie Chen
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
23
|
Zhang S, Yu J, Liu Y, Xiong B, Fang Y, Zhu Y, Li S, Sun L, Zhou B, Sun Y, Wang L, Yue W, Yin H, Xu H. Photosynthetic Bacteria-Hitchhiking 2D iMXene-mRNA Vaccine to Enable Photo-Immunogene Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307225. [PMID: 38742454 PMCID: PMC11267280 DOI: 10.1002/advs.202307225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/29/2024] [Indexed: 05/16/2024]
Abstract
Therapeutic mRNA vaccines have become powerful therapeutic tools for severe diseases, including infectious diseases and malignant neoplasms. mRNA vaccines encoding tumor-associated antigens provide unprecedented hope for many immunotherapies that have hit the bottleneck. However, the application of mRNA vaccines is limited because of biological instability, innate immunogenicity, and ineffective delivery in vivo. This study aims to construct a novel mRNA vaccine delivery nanosystem to successfully co-deliver a tumor-associated antigen (TAA) encoded by the Wilms' tumor 1 (WT1) mRNA. In this system, named PSB@Nb1.33C/mRNA, photosynthetic bacteria (PSB) efficiently delivers the iMXene-WT1 mRNA to the core tumor region using photo-driven and hypoxia-driven properties. The excellent photothermal therapeutic (PTT) properties of PSB and 2D iMxene (Nb1.33C) trigger tumor immunogenic cell death, which boosts the release of the WT1 mRNA. The released WT1 mRNA is translated, presenting the TAA and amplifying immune effect in vivo. The designed therapeutic strategy demonstrates an excellent ability to inhibit distant tumors and counteract postsurgical lung metastasis. Thus, this study provides an innovative and effective paradigm for tumor immunotherapy, i.e., photo-immunogene cancer therapy, and establishes an efficient delivery platform for mRNA vaccines, thereby opening a new path for the wide application of mRNA vaccines.
Collapse
Affiliation(s)
- Shen Zhang
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Jifeng Yu
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yunyun Liu
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalUltrasound Research and Education InstituteClinical Research Center for Interventional MedicineSchool of MedicineTongji University. Shanghai 200072P. R. China
- Department of Medical UltrasoundShanghai Tenth People's HospitalNanjing Medical UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Bing Xiong
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yan Fang
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalUltrasound Research and Education InstituteClinical Research Center for Interventional MedicineSchool of MedicineTongji University. Shanghai 200072P. R. China
- Department of Medical UltrasoundShanghai Tenth People's HospitalNanjing Medical UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Yuli Zhu
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Shaoyue Li
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalUltrasound Research and Education InstituteClinical Research Center for Interventional MedicineSchool of MedicineTongji University. Shanghai 200072P. R. China
- Department of Medical UltrasoundShanghai Tenth People's HospitalNanjing Medical UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Liping Sun
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalUltrasound Research and Education InstituteClinical Research Center for Interventional MedicineSchool of MedicineTongji University. Shanghai 200072P. R. China
- Department of Medical UltrasoundShanghai Tenth People's HospitalNanjing Medical UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Boyang Zhou
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yikang Sun
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Lifan Wang
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Wenwen Yue
- Department of Medical UltrasoundCenter of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalUltrasound Research and Education InstituteClinical Research Center for Interventional MedicineSchool of MedicineTongji University. Shanghai 200072P. R. China
- Department of Medical UltrasoundShanghai Tenth People's HospitalNanjing Medical UniversityShanghai200072P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentShanghai200072P. R. China
| | - Haohao Yin
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Huixiong Xu
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| |
Collapse
|
24
|
Jia J, Wang X, Lin X, Zhao Y. Engineered Microorganisms for Advancing Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313389. [PMID: 38485221 DOI: 10.1002/adma.202313389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Engineered microorganisms have attracted significant interest as a unique therapeutic platform in tumor treatment. Compared with conventional cancer treatment strategies, engineering microorganism-based systems provide various distinct advantages, such as the intrinsic capability in targeting tumors, their inherent immunogenicity, in situ production of antitumor agents, and multiple synergistic functions to fight against tumors. Herein, the design, preparation, and application of the engineered microorganisms for advanced tumor therapy are thoroughly reviewed. This review presents a comprehensive survey of innovative tumor therapeutic strategies based on a series of representative engineered microorganisms, including bacteria, viruses, microalgae, and fungi. Specifically, it offers extensive analyses of the design principles, engineering strategies, and tumor therapeutic mechanisms, as well as the advantages and limitations of different engineered microorganism-based systems. Finally, the current challenges and future research prospects in this field, which can inspire new ideas for the design of creative tumor therapy paradigms utilizing engineered microorganisms and facilitate their clinical applications, are discussed.
Collapse
Affiliation(s)
- Jinxuan Jia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaocheng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
25
|
Xiao T, Ma Y, Zhang Z, Zhang Y, Zhao Y, Zhou X, Wang X, Ge K, Guo J, Zhang J, Li Z, Liu H. Tailoring therapeutics via a systematic beneficial elements comparison between photosynthetic bacteria-derived OMVs and extruded nanovesicles. Bioact Mater 2024; 36:48-61. [PMID: 38434148 PMCID: PMC10904884 DOI: 10.1016/j.bioactmat.2024.02.025] [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: 11/16/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Photosynthetic bacteria (PSB) has shown significant potential as a drug or drug delivery system owing to their photothermal capabilities and antioxidant properties. Nevertheless, the actualization of their potential is impeded by inherent constraints, including their considerable size, heightened immunogenicity and compromised biosafety. Conquering these obstacles and pursuing more effective solutions remains a top priority. Similar to extracellular vesicles, bacterial outer membrane vesicles (OMVs) have demonstrated a great potential in biomedical applications. OMVs from PSB encapsulate a rich array of bioactive constituents, including proteins, nucleic acids, and lipids inherited from their parent cells. Consequently, they emerge as a promising and practical alternative. Unfortunately, OMVs have suffered from low yield and inconsistent particle sizes. In response, bacteria-derived nanovesicles (BNVs), created through controlled extrusion, adeptly overcome the challenges associated with OMVs. However, the differences, both in composition and subsequent biological effects, between OMVs and BNVs remain enigmatic. In a groundbreaking endeavor, our study meticulously cultivates PSB-derived OMVs and BNVs, dissecting their nuances. Despite minimal differences in morphology and size between PSB-derived OMVs and BNVs, the latter contains a higher concentration of active ingredients and metabolites. Particularly noteworthy is the elevated levels of lysophosphatidylcholine (LPC) found in BNVs, known for its ability to enhance cell proliferation and initiate downstream signaling pathways that promote angiogenesis and epithelialization. Importantly, our results indicate that BNVs can accelerate wound closure more effectively by orchestrating a harmonious balance of cell proliferation and migration within NIH-3T3 cells, while also activating the EGFR/AKT/PI3K pathway. In contrast, OMVs have a pronounced aptitude in anti-cancer efforts, driving macrophages toward the M1 phenotype and promoting the release of inflammatory cytokines. Thus, our findings not only provide a promising methodological framework but also establish a definitive criterion for discerning the optimal application of OMVs and BNVs in addressing a wide range of medical conditions.
Collapse
Affiliation(s)
- Tingshan Xiao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Yichuan Ma
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
- College of Chemistry & Materials Science, Hebei University, Baoding, 071002, China
| | - Ziyang Zhang
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Yixin Zhang
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Yu Zhao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xiaohan Zhou
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523000, China
| | - Xueyi Wang
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523000, China
| | - Kun Ge
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
- College of Chemistry & Materials Science, Hebei University, Baoding, 071002, China
| | - Junshu Guo
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523000, China
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
- College of Chemistry & Materials Science, Hebei University, Baoding, 071002, China
| | - Zhenhua Li
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523000, China
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| |
Collapse
|
26
|
Zhou Y, Li Q, Wu Y, Zhang W, Ding L, Ji C, Li P, Chen T, Feng L, Tang BZ, Huang X. Synergistic Brilliance: Engineered Bacteria and Nanomedicine Unite in Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313953. [PMID: 38400833 DOI: 10.1002/adma.202313953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Engineered bacteria are widely used in cancer treatment because live facultative/obligate anaerobes can selectively proliferate at tumor sites and reach hypoxic regions, thereby causing nutritional competition, enhancing immune responses, and producing anticancer microbial agents in situ to suppress tumor growth. Despite the unique advantages of bacteria-based cancer biotherapy, the insufficient treatment efficiency limits its application in the complete ablation of malignant tumors. The combination of nanomedicine and engineered bacteria has attracted increasing attention owing to their striking synergistic effects in cancer treatment. Engineered bacteria that function as natural vehicles can effectively deliver nanomedicines to tumor sites. Moreover, bacteria provide an opportunity to enhance nanomedicines by modulating the TME and producing substrates to support nanomedicine-mediated anticancer reactions. Nanomedicine exhibits excellent optical, magnetic, acoustic, and catalytic properties, and plays an important role in promoting bacteria-mediated biotherapies. The synergistic anticancer effects of engineered bacteria and nanomedicines in cancer therapy are comprehensively summarized in this review. Attention is paid not only to the fabrication of nanobiohybrid composites, but also to the interpromotion mechanism between engineered bacteria and nanomedicine in cancer therapy. Additionally, recent advances in engineered bacteria-synergized multimodal cancer therapies are highlighted.
Collapse
Affiliation(s)
- Yaofeng Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Qianying Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Wan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Lu Ding
- Department of Cardiology, Jiangxi Hypertension Research Institute, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, 310030, P. R. China
| | - Ping Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330036, P. R. China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ben Zhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, P. R. China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| |
Collapse
|
27
|
Fu Y, Li J, Cai W, Huang Y, Liu X, Ma Z, Tang Z, Bian X, Zheng J, Jiang J, Li C. The emerging tumor microbe microenvironment: From delineation to multidisciplinary approach-based interventions. Acta Pharm Sin B 2024; 14:1560-1591. [PMID: 38572104 PMCID: PMC10985043 DOI: 10.1016/j.apsb.2023.11.018] [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: 08/28/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024] Open
Abstract
Intratumoral microbiota has become research hotspots, and emerges as a non-negligent new component of tumor microenvironments (TME), due to its powerful influence on tumor initiation, metastasis, immunosurveillance and prognosis despite in low-biomass. The accumulations of microbes, and their related components and metabolites within tumor tissues, endow TME with additional pluralistic features which are distinct from the conventional one. Therefore, it's definitely necessary to comprehensively delineate the sophisticated landscapes of tumor microbe microenvironment, as well as their functions and related underlying mechanisms. Herein, in this review, we focused on the fields of tumor microbe microenvironment, including the heterogeneity of intratumor microbiota in different types of tumors, the controversial roles of intratumoral microbiota, the basic features of tumor microbe microenvironment (i.e., pathogen-associated molecular patterns (PAMPs), typical microbial metabolites, autophagy, inflammation, multi-faceted immunomodulation and chemoresistance), as well as the multidisciplinary approach-based intervention of tumor microbiome for cancer therapy by applying wild-type or engineered live microbes, microbiota metabolites, antibiotics, synthetic biology and rationally designed biomaterials. We hope our work will provide valuable insight to deeply understand the interplay of cancer-immune-microbial, and facilitate the development of microbes-based tumor-specific treatments.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jia Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Wenyun Cai
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
28
|
Zhang JA, Haddleton D, Wilson P, Zhu LH, Dai CY, Zhao LL. pH-Responsive Amphiphilic Triblock Fluoropolymers as Assemble Oxygen Nanoshuttles for Enhancing PDT against Hypoxic Tumor. Bioconjug Chem 2024; 35:400-411. [PMID: 38366969 DOI: 10.1021/acs.bioconjchem.4c00029] [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: 02/19/2024]
Abstract
Photodynamic therapy (PDT) is a cancer treatment strategy that utilizes photosensitizers to convert oxygen within tumors into reactive singlet oxygen (1O2) to lyse tumor cells. Nevertheless, pre-existing tumor hypoxia and oxygen consumption during PDT can lead to an insufficient oxygen supply, potentially reducing the photodynamic efficacy. In response to this issue, we have devised a pH-responsive amphiphilic triblock fluorinated polymer (PDP) using copper-mediated RDRP. This polymer, composed of poly(ethylene glycol) methyl ether acrylate, 2-(diethylamino)ethyl methacrylate, and (perfluorooctyl)ethyl acrylate, self-assembles in an aqueous environment. Oxygen, chlorine e6 (Ce6), and doxorubicin (DOX) can be codelivered efficiently by PDP. The incorporation of perfluorocarbon into the formulation enhances the oxygen-carrying capacity of PDP, consequently extending the lifetime of 1O2. This increased lifetime, in turn, amplifies the PDT effect and escalates the cellular cytotoxicity. Compared with PDT alone, PDP@Ce6-DOX-O2 NPs demonstrated significant inhibition of tumor growth. This study proposes a novel strategy for enhancing the efficacy of PDT.
Collapse
Affiliation(s)
- Jun-An Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - David Haddleton
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Paul Wilson
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Lin-Hua Zhu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Key Laboratory of Functional Organic Polymers of Haikou, Tropical Functional Polymer Materials Engineering Research Center of Hainan, Haikou 571158, China
| | - Chun-Yan Dai
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Key Laboratory of Functional Organic Polymers of Haikou, Tropical Functional Polymer Materials Engineering Research Center of Hainan, Haikou 571158, China
| | - Lin-Lu Zhao
- College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
29
|
Han D, Wang F, Ma Y, Zhao Y, Zhang W, Zhang Z, Liu H, Yang X, Zhang C, Zhang J, Li Z. Redirecting Antigens by Engineered Photosynthetic Bacteria and Derived Outer Membrane Vesicles for Enhanced Cancer Immunotherapy. ACS NANO 2023; 17:18716-18731. [PMID: 37782086 DOI: 10.1021/acsnano.3c01912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Significant strides have been made in the development of cancer vaccines to combat malignant tumors. However, the natural immunosuppressive environment within tumors, known as the tumor microenvironment (TME), hampers the uptake and presentation of antigens by antigen-presenting cells (APCs) within the tumor itself. This limitation results in inadequate activation of immune responses against cancer. In contrast, immune cells in peritumoral tissue maintain their normal functions. In this context, we present an interesting approach to enhance cancer immunotherapy by utilizing engineered photosynthetic bacteria (PSB) and their outer membrane vesicles (OMVPSB) to capture and transport antigens to the outer regions of the tumor. We modified PSB with maleimide (PSB-MAL), which, when exposed to near-infrared (NIR) laser-mediated photothermal therapy (PTT), induced extensive cancer cell death and the release of tumor antigens. Subsequently, the NIR-phototactic PSB-MAL transported these tumor antigens to the peripheral regions of the tumor under NIR laser exposure. Even more intriguingly, PSB-MAL-derived OMVPSB-MAL effectively captured and delivered antigens to tumor-draining lymph nodes (TDLNs). This facilitated enhanced antigen presentation by mature and fully functional APCs in the TDLNs. This intricate communication network between PSB-MAL, the OMVPSB-MAL, and APCs promoted the efficient presentation of tumor antigens in the tumor periphery and TDLNs. Consequently, there was a notable increase in the infiltration of cytotoxic T lymphocytes (CTLs) into the tumor, triggering potent antitumor immune responses in both melanoma and breast cancer models. This cascade of events resulted in enhanced suppression of tumor metastasis and recurrence, underscoring the robust efficacy of our approach. Our interesting study, harnessing the potential of bacteria and OMVs to redirect tumor antigens for enhanced cancer immunotherapy, provides a promising path toward the development of personalized cancer vaccination strategies.
Collapse
Affiliation(s)
- Dandan Han
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong 510515, China
| | - Fei Wang
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong 510515, China
| | - Yichuan Ma
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Yu Zhao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Wei Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Ziyang Zhang
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Xinjian Yang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Chi Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Zhenhua Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong 510515, China
| |
Collapse
|
30
|
Luo W, Zhang Z, Zhou D, Jiang Y, Yang J, He B, Yu H, Song Y. Deep Tumor Penetration of CRISPR-Cas System for Photothermal-Sensitized Immunotherapy via Probiotics. NANO LETTERS 2023; 23:8081-8090. [PMID: 37615340 DOI: 10.1021/acs.nanolett.3c02061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Since central cells are more malignant and aggressive in solid tumors, improving penetration of therapeutic agents and activating immunity in tumor centers exhibit great potential in cancer therapies. Here, polydopamine-coated Escherichia coli Nissle 1917 (EcN) bearing CRISPR-Cas9 plasmid-loaded liposomes (Lipo-P) are applied for enhanced immunotherapy in deep tumors through activation of innate and adaptive immunity simultaneously. After accumulation in the tumor center through hypoxia targeting, Lipo-P could be detached under the reduction of reactive oxygen species (ROS)-responsive linkers, lowering the thermal resistance of cancer cells via Hsp90α depletion. Owing to that, heating induced by polydopamine upon near-infrared irradiation could achieve effective tumor ablation. Furthermore, mild photothermal therapy induces immunogenic cell death, as bacterial infections in tumor tissues trigger innate immunity. This bacteria-assisted approach provides a promising photothermal-sensitized immunotherapy in deep tumors.
Collapse
Affiliation(s)
- Wen Luo
- Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Zhibin Zhang
- Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Dongtao Zhou
- Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Yateng Jiang
- Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, 210006, Nanjing, China
| | - Haijia Yu
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yujun Song
- Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
31
|
Lu Q, Liu T, Han Z, Zhao J, Fan X, Wang H, Song J, Ye H, Sun J. Revolutionizing cancer treatment: The power of cell-based drug delivery systems. J Control Release 2023; 361:604-620. [PMID: 37579974 DOI: 10.1016/j.jconrel.2023.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/30/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Intravenous administration of drugs is a widely used cancer therapy approach. However, the efficacy of these drugs is often hindered by various biological barriers, including circulation, accumulation, and penetration, resulting in poor delivery to solid tumors. Recently, cell-based drug delivery platforms have emerged as promising solutions to overcome these limitations. These platforms offer several advantages, including prolonged circulation time, active targeting, controlled release, and excellent biocompatibility. Cell-based delivery systems encompass cell membrane coating, intracellular loading, and extracellular backpacking. These innovative platforms hold the potential to revolutionize cancer diagnosis, monitoring, and treatment, presenting a plethora of opportunities for the advancement and integration of pharmaceuticals, medicine, and materials science. Nevertheless, several technological, ethical, and financial barriers must be addressed to facilitate the translation of these platforms into clinical practice. In this review, we explore the emerging strategies to overcome these challenges, focusing specifically on the functions and advantages of cell-mediated drug delivery in cancer treatment.
Collapse
Affiliation(s)
- Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tian Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zeyu Han
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jian Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xiaoyuan Fan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Helin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jiaxuan Song
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China; Multi-Scale Robotics Lab (MSRL), Institute of Robotics & Intelligent Systems (IRIS), ETH Zurich, Zurich 8092, Switzerland.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
32
|
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: 22] [Impact Index Per Article: 11.0] [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
|
33
|
Wang H, Shang J, Yang F, Zhang S, Cui J, Hou X, Li Y, Liu W, Shu X, Liu Y, Sun B. Modulation of tumour hypoxia by ultrasound-responsive microbubbles to enhance the sono-photodynamic therapy effect on triple-negative breast cancer. Photodiagnosis Photodyn Ther 2023:103642. [PMID: 37271488 DOI: 10.1016/j.pdpdt.2023.103642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Sono-photodynamic therapy (SPDT) is an oxidative stress-dependent antitumour treatment modality. Due to the hypoxic tumour microenvironment, the antitumour effect of SPDT is limited. In this study, we developed lipid vesicles to transport a photosensitizer (chlorin e6, Ce6) and oxygen into tumours to promote SPDT efficiency on triple-negative breast cancer in vitro and in vivo. The results showed that compared with the same concentration of free Ce6, Lipo-Ce6 produced a higher singlet oxygen level under light irradiation. Cellular Lipo-Ce6 accumulation was 4-fold higher than that of free Ce6. The cytotoxicity on 4T1 cells caused by Lipo-Ce6-SPDT was significantly stronger than that caused by free Ce6-SPDT, and oxygen microbubbles (O2MB) further enhanced the cytotoxicity of Lipo-Ce6-SPDT under hypoxic conditions. Cellular ROS production in the Lipo-Ce6-SPDT+O2MB group was approximately 2.5-fold higher than that in the Lipo-Ce6-SPDT+C3F8MB group. Furthermore, O2MB rapidly relieved 4T1 subcutaneous xenograft hypoxia conditions under ultrasound exposure and significantly improved the antitumour activity of SPDT in vivo. These results indicate that the combination of O2MB and a high-activity liposome photosensitizer can significantly enhance the antitumour efficiency of SPDT for hypoxic tumours.
Collapse
Affiliation(s)
- Haiping Wang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China
| | - Jinting Shang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China; Huazhong University of Science and Technology; Advanced Technology Institute of Suzhou Chinese Academy of Science,Co.,Ltd
| | - Fang Yang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China
| | - Song Zhang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Jingsong Cui
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xiaoying Hou
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, Guangxi, China
| | - Wei Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xiji Shu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Yuchen Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China.
| | - Binlian Sun
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China.
| |
Collapse
|
34
|
Wang XM, Pan S, Chen L, Wang L, Dai YT, Luo T, Li WW. Biogenic Copper Selenide Nanoparticles for Near-Infrared Photothermal Therapy Application. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37262434 DOI: 10.1021/acsami.3c03611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Near-infrared (NIR) photothermal therapy (PTT) is attractive for cancer treatment but is currently restricted by limited availability and insufficient NIR-II photoactivity of photothermal agents, for which artificial nanomaterials are usually used. Here, we report the first use of biogenic nanomaterials for PTT application. A fine-controlled extracellular biosynthesis of copper selenide nanoparticles (bio-Cu2-xSe) by Shewanella oneidensis MR-1 was realized. The resulting bio-Cu2-xSe, with fine sizes (∼35.5 nm) and high product purity, exhibited 76.9% photothermal conversion efficiency under 1064 nm laser irradiation, outperforming almost all the existing counterparts. The protein capping also imparted good biocompatibility to bio-Cu2-xSe to favor a safe PTT application. The in vivo PTT with injected bio-Cu2-xSe in mice (without extraction nor further modification) showed 87% tumor ablation without impairing the normal organisms. Our work not only opens a green route to synthesize the NIR-II photothermal nanomaterial but may also lay a basis for the development of bacteria-nanomaterial hybrid therapy technologies.
Collapse
Affiliation(s)
- Xue-Meng Wang
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
- Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, School of Environmental and Municipal Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, China
| | - Shaoshan Pan
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230026, China
| | - Lin Chen
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Li Wang
- School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230026, China
| | - Yi-Tao Dai
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230026, China
| | - Wen-Wei Li
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| |
Collapse
|
35
|
Wang T, Yin Q, Huang HY, Wang Z, Song H, Luo X. Probiotic Escherichia coli Nissle 1917 propelled micro-robot with pH sensitivity for hypoxia targeted intestinal tumor therapy. Colloids Surf B Biointerfaces 2023; 225:113277. [PMID: 36996630 DOI: 10.1016/j.colsurfb.2023.113277] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Poor drug penetration in hypoxia area of solid tumor is a big challenge for intestinal tumor therapy and thus it is crucial to develop an effective strategy to overcome this challenge. Compared with other bacteria used for construction of hypoxia targeted bacteria micro-robot, the Escherichia coli Nissle 1917 (EcN) bacteria are nonpathogenic Gram-negative probiotic and can especially target and identify the signal molecules in the hypoxic region of tumor, and thus, in this study, we choose EcN to construct a bacteria propelled micro-robot for targeting intestinal tumor therapy. Firstly, the MSNs@DOX with average diameter of 200 nm were synthesized and conjugated with EcN bacteria using EDC/NHS chemical crosslinking method to construct a EcN propelled micro-robot. The motility of micro-robot was then evaluated and the motion velocity of EcN-pMSNs@DOX was 3.78 µm/s. Compared with pMSNs@DOX without EcN driven, EcN bacteria propelled micro-robot transported much more pMSNs@DOX into the inner of HCT-116 3D multicellular tumor spheroids. However, the EcN bacteria are non-intracelluar bacteria which lead to the micro-robot can not directly enter into tumor cells. Therefore, we utilized acid-labile linkers of cis-aconitic amido bone to link EcN with MSNs@DOX nanoparticles to achieve the pH sensitive separation of EcN with MSNs@DOX from the micro-robot. At 4 h of incubation, the isolated MSNs@DOX began to enter into the tumor cells through CLSM observation. In vitro live/dead staining results show that EcN-pMSNs@DOX induced much more cell death than pMSNs@DOX at 24 and 48 h of incubation with HCT-116 tumor cells in acid culture media (pH 5.3). For the validation of the therapeutic efficacy of the micro-robot for intestinal tumor, we established the HCT-116 subcutaneous transplantation tumor model. After 28 days of treatment, EcN-pMSNs@DOX dramatically inhibit tumor growth with tumor volume was around 689 mm3, induce much more tumor tissues necrosis and apoptosis. Finally, the toxicity of this micro-robot was investigated by pathological analysis the liver and heart tissues. We expect that the pH sensitive EcN propelled micro-robot here we constructed may be a safe and feasible strategy for intestinal tumor therapy.
Collapse
|
36
|
Wang X, Sun Y, Wangpraseurt D. Engineered photoresponsive biohybrids for tumor therapy. SMART MEDICINE 2023; 2:e20220041. [PMID: 39188274 PMCID: PMC11235730 DOI: 10.1002/smmd.20220041] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 08/28/2024]
Abstract
Engineered biohybrids have recently emerged as innovative biomimetic platforms for cancer therapeutic applications. Particularly, engineered photoresponsive biohybrids hold tremendous potential against tumors due to their intriguing biomimetic properties, photoresponsive ability, and enhanced biotherapeutic functions. In this review, the design principles of engineered photoresponsive biohybrids and their latest progresses for tumor therapy are summarized. Representative engineered photoresponsive biohybrids are highlighted including biomolecules-associated, cell membrane-based, eukaryotic cell-based, bacteria-based, and algae-based photoresponsive biohybrids. Representative tumor therapeutic modalities of the engineered photoresponsive biohybrids are presented, including photothermal therapy, photodynamic therapy, synergistic therapy, and tumor therapy combined with tissue regeneration. Moreover, the challenges and future perspectives of these photoresponsive biohybrids for clinical practice are discussed.
Collapse
Affiliation(s)
- Xiaocheng Wang
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Yazhi Sun
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Daniel Wangpraseurt
- Department of NanoEngineeringUniversity of California San DiegoSan DiegoCaliforniaUSA
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCaliforniaUSA
| |
Collapse
|
37
|
Xu Y, Wu Y, Hu Y, Xu M, Liu Y, Ding Y, Chen J, Huang X, Wen L, Li J, Zhu C. Bacteria-based multiplex system eradicates recurrent infections with drug-resistant bacteria via photothermal killing and protective immunity elicitation. Biomater Res 2023; 27:27. [PMID: 37024953 PMCID: PMC10080897 DOI: 10.1186/s40824-023-00363-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND The high mortality associated with drug-resistant bacterial infections is an intractable clinical problem resulting from the low susceptibility of these bacteria to antibiotics and the high incidence of recurrent infections. METHODS Herein, a photosynthetic bacteria-based multiplex system (Rp@Al) composed of natural Rhodopseudomonas palustris (Rp) and Food and Drug Administration-approved aluminum (Al) adjuvant, was developed to combat drug-resistant bacterial infections and prevent their recurrence. We examined its photothermal performance and in vitro and in vivo antibacterial ability; revealed its protective immunomodulatory effect; verified its preventative effect on recurrent infections; and demonstrated the system's safety. RESULTS Rp@Al exhibits excellent photothermal properties with an effective elimination of methicillin-resistant Staphylococcus aureus (MRSA). In addition, Rp@Al enhances dendritic cell activation and further triggers a T helper 1 (TH1)/TH2 immune response, resulting in pathogen-specific immunological memory against recurrent MRSA infection. Upon second infection, Rp@Al-treated mice show significantly lower bacterial burden, faster abscess recovery, and higher survival under near-lethal infection doses than control mice. CONCLUSIONS This innovative multiplex system, with superior photothermal and immunomodulatory effects, presents great potential for the treatment and prevention of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Youcui Xu
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yi Wu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yi Hu
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Mengran Xu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yanyan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yuting Ding
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Jing Chen
- School of Life Sciences, Hefei Normal University, Hefei, 230601, Anhui, China
| | - Xiaowan Huang
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Longping Wen
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Chen Zhu
- Department of Orthopaedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
38
|
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
|
39
|
Guo H, Cao Z, Li J, Fu Z, Lin S, Wang L, Liu J. Integrating Bacteria with a Ternary Combination of Photosensitizers for Monochromatic Irradiation-Mediated Photoacoustic Imaging-Guided Synergistic Photothermal Therapy. ACS NANO 2023; 17:5059-5071. [PMID: 36847803 DOI: 10.1021/acsnano.3c00032] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Photosensitizer-based therapy often suffers from unitary and easily attenuated photosensitive effects, limited tumor penetration and retention, and requirement of multiple irradiation for combination therapy, which largely restrict its application. Here, bacteria are integrated with a monochromatic irradiation-mediated ternary combination of photosensitizers for photoacoustic imaging-guided synergistic photothermal therapy. Bacteria that are bioengineered to express natural melanin are decorated with dual synthetic photosensitizers by nanodeposition with indocyanine green and polydopamine under a cytocompatible condition. The combined photosensitizers, which share an adequate excitation at 808 nm, endow integrated bacteria with a stable triple photoacoustic and photothermal effect under a monochromatic irradiation. Due to their living characteristics, these bacteria preferentially colonize hypoxic tumor tissue with homogeneous distribution and durable retention and generate uniform imaging signals and a sufficient heating of tumor upon laser irradiation. Supported by significantly inhibited tumor growth and extended survival of animals in different tumor-bearing murine models, our work proposes the development of bacteria-based innovative photosensitizers for imaging-guided therapy.
Collapse
Affiliation(s)
- Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhenzhen Fu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
40
|
Debasmita D, Ghosh SS, Chattopadhyay A. Living Gut Bacteria Functionalized with Gold Nanoclusters and Drug for Facile Cancer Theranostics. ACS APPLIED BIO MATERIALS 2023; 6:628-639. [PMID: 36651899 DOI: 10.1021/acsabm.2c00911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bacbots are potent self-propelling vehicles for targeted therapy that can be guided by chemical and biochemical stimuli of the host. In addition, they can be guided externally by the use of magnetic field or other physical forces. The challenge is to incorporate drugs and diagnostic tools in living bacteria with retention of theranostic activity until reaching the targets and easy clearance of the remainder following the treatment. We report that living Lactobacillus rhamnosus, when functionalized with photoluminescent Au nanoclusters and the anticancer drug methotrexate, was cytotoxic to monolayer and spheroids of cancer cells (HeLa and HT29) even at a low dose of bacteria used (107 cfu/mL). The observed cell death was nearly 90% in HeLa spheroids and 70% in HT29 spheroids. Further, functionalization of the bacterial surface with the nanoclusters helped incorporate the drug onto their cell surfaces. The drug and nanocluster-loaded bacteria annihilated the cells and the spheroids in a rather short time (6 h) that revealed the specificity and effectiveness of the bacbots. The bacbots exhibited synergistic toxicity on the cells as their effect was more than the drug and the bacteria individually. This higher toxicity could be associated with elevated levels of reactive oxygen species generated in the bacbot-treated cells. The multifunctional bacbots reported here provide an option for guided therapy with the natural variant of the human gut-friendly living bacteria without the need for attenuation or genetic modification.
Collapse
|
41
|
Zhuang Y, Liu K, He Q, Gu X, Jiang C, Wu J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e203. [PMID: 36703877 PMCID: PMC9870816 DOI: 10.1002/mco2.203] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoxia is a persistent physiological feature of many different solid tumors and a key driver of malignancy, and in recent years, it has been recognized as an important target for cancer therapy. Hypoxia occurs in the majority of solid tumors due to a poor vascular oxygen supply that is not sufficient to meet the needs of rapidly proliferating cancer cells. A hypoxic tumor microenvironment (TME) can reduce the effectiveness of other tumor therapies, such as radiotherapy, chemotherapy, and immunotherapy. In this review, we discuss the critical role of hypoxia in tumor development, including tumor metabolism, tumor immunity, and tumor angiogenesis. The treatment methods for hypoxic TME are summarized, including hypoxia-targeted therapy and improving oxygenation by alleviating tumor hypoxia itself. Hyperoxia therapy can be used to improve tissue oxygen partial pressure and relieve tumor hypoxia. We focus on the underlying mechanisms of hyperoxia and their impact on current cancer therapies and discuss the prospects of hyperoxia therapy in cancer treatment.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Kua Liu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Qinyu He
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Xiaosong Gu
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| |
Collapse
|
42
|
Prospects for hypoxia-based drug delivery platforms for the elimination of advanced metastatic tumors: From 3D modeling to clinical concepts. J Control Release 2023; 353:1002-1022. [PMID: 36516901 DOI: 10.1016/j.jconrel.2022.12.009] [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: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia is a unique characteristic of the solid tumor microenvironment. Hypoxia contributes to multi-drug resistance, metastasis and cancer relapse through numerous molecular pathways, but at the same time provides an opportunity for the development of novel drugs or modalities specifically targeting hypoxic tumor regions. Given the high significance of tumor hypoxia in therapeutic results, we here discuss a variety of hypoxia-adopted strategies, and their potential and utility in the treatment of deep-seated hypoxic tumor cells. We discuss the merits and demerits of these approaches, as well as their combination with other approaches such as photodynamic therapy. We also survey the currently available 3D hypoxia modeling systems, in particular organoid-based microfluidics. Finally, we discuss the potential and the current status of preclinical tumor hypoxia approaches in clinical trials for advanced cancer. We believe that multi-modal imaging and therapeutic hypoxia adopted drug delivery platforms could provide better efficacy and safety profiles, and more importantly personalized therapy. Determining the hypoxia status of tumors could offer a second chance for the clinical translation of hypoxia-based agents, such as hypoxia activated prodrugs (HAPs) from bench to bedside.
Collapse
|
43
|
Shi H, Chen L, Liu Y, Wen Q, Lin S, Wen Q, Lu Y, Dai J, Li J, Xiao S, Fu S. Bacteria-Driven Tumor Microenvironment-Sensitive Nanoparticles Targeting Hypoxic Regions Enhances the Chemotherapy Outcome of Lung Cancer. Int J Nanomedicine 2023; 18:1299-1315. [PMID: 36945255 PMCID: PMC10024911 DOI: 10.2147/ijn.s396863] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/29/2023] [Indexed: 03/17/2023] Open
Abstract
Background Chemotherapy still plays a dominant role in cancer treatment. However, the inability of conventional chemotherapeutic drugs to reach the hypoxic zone of solid tumors significantly weakens their efficacy. Bacteria-mediated drug delivery systems can be an effective targeting strategy for improving the therapeutic outcomes in cancer. Anaerobic bacteria have the unique ability to selectively transport drug loads to the hypoxic regions of tumors. Methods We designed a Bifidobacterium infantis (Bif)-based biohybrid (Bif@PDA-PTX-NPs) to deliver polydopamine (PDA)-coated paclitaxel nanoparticles (PTX-NPs) to tumor tissues. Results The self-driven Bif@PDA-PTX-NPs maintained the toxicity of PTX as well as the hypoxic homing tendency of Bif. Furthermore, Bif@PDA-PTX-NPs significantly inhibited the growth of A549 xenografts in nude mice, and prolonged the survival of the tumor-bearing mice compared to the other PTX formulations without any systemic or localized toxicity. Conclusion The Bif@PDA-PTX-NPs biohybrids provide a new therapeutic strategy for targeted chemotherapy to solid tumors.
Collapse
Affiliation(s)
- Huan Shi
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Lan Chen
- Department of Oncology, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Yanlin Liu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Qinglian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Sheng Lin
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Qian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Yun Lu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Jie Dai
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Jianmei Li
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Susu Xiao
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Shaozhi Fu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Correspondence: Shaozhi Fu, Tel +86 830-3165698, Fax +86 830-3165690, Email
| |
Collapse
|
44
|
Liu S, Fang L, Ding H, Zhang Y, Li W, Liu B, Dong S, Tian B, Feng L, Yang P. Alternative Strategy to Optimize Cerium Oxide for Enhanced X-ray-Induced Photodynamic Therapy. ACS NANO 2022; 16:20805-20819. [PMID: 36378717 DOI: 10.1021/acsnano.2c08047] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The emergence of X-ray-induced photodynamic therapy (X-PDT) holds tremendous promise for clinical deep-penetrating cancer therapy. However, the clinical application of X-PDT in cancer treatment is still limited due to the hypoxic property of cancerous tissue, the inherent antioxidant system of tumor cells, and the difficulty in matching the absorption spectra of photosensitizers. Herein, a versatile core-shell radiosensitizer (SCNPs@DMSN@CeOx-PEG, denoted as SSCP) was elaborately designed and constructed to enhance X-PDT by coating tunable mesoporous silica on nanoscintillators, followed by embedding the cerium oxide nanoparticles in situ. The obtained SSCP radiosensitizer demonstrated a distinct blue-shift in the ultraviolet light region, so that it could perfectly absorb the ultraviolet light converted by the SCNPs core, resulting in the formation of photoinduced electron-hole (e--h+) pairs separation to generate reactive oxygen species (ROS). In addition, the cerium oxide exhibits high glutathione consumption to heighten ROS accumulation, and catalase-like activity to alleviate the hypoxia, which further enhances the efficiency of radiotherapy. Benefiting from the abundant Lu and Ce elements, the computed tomography imaging performance of SSCP is about 3.79-fold that of the clinical contrast agent (iohexol), which has great potential in both preclinical imaging and clinical translation.
Collapse
Affiliation(s)
- Shikai Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Linyang Fang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - He Ding
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Yangyang Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Wenting Li
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Bin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Shuming Dong
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Boshi Tian
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, Harbin Engineering University, Harbin 150001, People's Republic of China
| |
Collapse
|
45
|
Han D, Zhang X, Ma Y, Yang X, Li Z. The development of live microorganism-based oxygen shuttles for enhanced hypoxic tumor therapy. Mater Today Bio 2022; 18:100517. [PMID: 36578285 PMCID: PMC9791452 DOI: 10.1016/j.mtbio.2022.100517] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Hypoxia is a prominent feature of malignant tumors and contributes to tumor proliferation, metastasis, and drug resistance in various solid tumors. Therefore, improving tumor oxygenation is crucial for curing tumors. To date, multiple strategies, including oxygen delivering and producing materials, have been designed to increase the oxygen concentration in hypoxic tumors. However, the unsustainable supply of oxygen is still the main obstacle, resulting in a suboptimal outcome in treating oxygen-deprived tumors. Thus, a sufficient oxygen supply is highly desirable in the treatment of hypoxic tumors. Photosynthesis, as the main source of oxygen in nature through the conversion of light energy into chemical energy and oxygen, has been widely studied in scientific research. Moreover, photosynthetic microorganisms have been increasingly applied in cancer therapy by increasing oxygenation, which improves the therapeutic effect of oxygen-consuming tumor therapeutic tools such as radiotherapy and photodynamic therapy. In this review, we summarize recent advances in the design and manufacture of live bacteria as oxygen shuttles for a new generation of hypoxic tumor treatment strategies. Finally, current challenges and future directions are also discussed for successfully addressing hypoxic tumor issues.
Collapse
Affiliation(s)
- Dandan Han
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China,College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xing Zhang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Yichuan Ma
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xinjian Yang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China,Corresponding author.
| | - Zhenhua Li
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangdong, 510515, PR China,Corresponding author. Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China.
| |
Collapse
|
46
|
Liang S, Wang C, Shao Y, Wang Y, Xing D, Geng Z. Recent advances in bacteria-mediated cancer therapy. Front Bioeng Biotechnol 2022; 10:1026248. [PMID: 36312554 PMCID: PMC9597243 DOI: 10.3389/fbioe.2022.1026248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Cancer is among the leading cause of deaths worldwide. Although conventional therapies have been applied in the fight against the cancer, the poor oxygen, low extracellular pH, and high interstitial fluid pressure of the tumor microenvironment mean that these treatments fail to completely eradicate cancer cells. Recently, bacteria have increasingly been considered to be a promising platform for cancer therapy thanks to their many unique properties, such as specific tumor-targeting ability, high motility, immunogenicity, and their use as gene or drug carriers. Several types of bacteria have already been used for solid and metastatic tumor therapies, with promising results. With the development of synthetic biology, engineered bacteria have been endowed with the controllable expression of therapeutic proteins. Meanwhile, nanomaterials have been widely used to modify bacteria for targeted drug delivery, photothermal therapy, magnetothermal therapy, and photodynamic therapy, while promoting the antitumor efficiency of synergistic cancer therapies. This review will provide a brief introduction to the foundation of bacterial biotherapy. We begin by summarizing the recent advances in the use of many different types of bacteria in multiple targeted tumor therapies. We will then discuss the future prospects of bacteria-mediated cancer therapies.
Collapse
Affiliation(s)
- Shuya Liang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Wang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingchun Shao
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanhong Wang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Yanhong Wang, ; Dongming Xing, ; Zhongmin Geng,
| | - Dongming Xing
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
- *Correspondence: Yanhong Wang, ; Dongming Xing, ; Zhongmin Geng,
| | - Zhongmin Geng
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Yanhong Wang, ; Dongming Xing, ; Zhongmin Geng,
| |
Collapse
|
47
|
Chen W, He C, Qiao N, Guo Z, Hu S, Song Y, Wang H, Zhang Z, Ke B, Sun X. Dual drugs decorated bacteria irradiate deep hypoxic tumor and arouse strong immune responses. Biomaterials 2022; 286:121582. [DOI: 10.1016/j.biomaterials.2022.121582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023]
|
48
|
Yan H, Fan M, Liu H, Xiao T, Han D, Che R, Zhang W, Zhou X, Wang J, Zhang C, Yang X, Zhang J, Li Z. Microbial hydrogen "manufactory" for enhanced gas therapy and self-activated immunotherapy via reduced immune escape. J Nanobiotechnology 2022; 20:280. [PMID: 35705974 PMCID: PMC9199139 DOI: 10.1186/s12951-022-01440-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As an antioxidant, hydrogen (H2) can selectively react with the highly toxic hydroxyl radical (·OH) in tumor cells to break the balance of reactive oxygen species (ROS) and cause oxidative stress. However, due to the high diffusibility and storage difficulty of hydrogen, it is impossible to achieve long-term release at the tumor site, which highly limited their therapeutic effect. RESULTS Photosynthetic bacteria (PSB) release a large amount of hydrogen to break the balance of oxidative stress. In addition, as a nontoxic bacterium, PSB could stimulate the immune response and increase the infiltration of CD4+ and CD8+ T cells. More interestingly, we found that hydrogen therapy induced by our live PSB did not lead to the up-regulation of PD-L1 after stimulating the immune response, which could avoid the tumor immune escape. CONCLUSION Hydrogen-immunotherapy significantly kills tumor cells. We believe that our live microbial hydrogen production system provides a new strategy for cancer hydrogen treatment combining with enhanced immunotherapy without up-regulating PD-L1.
Collapse
Affiliation(s)
- Hongyu Yan
- Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Miao Fan
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Tingshan Xiao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Dandan Han
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Ruijun Che
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Wei Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth Peoples' Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Xiaohan Zhou
- Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China
| | - June Wang
- Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China
| | - Chi Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth Peoples' Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Xinjian Yang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, People's Republic of China.
| | - Zhenhua Li
- Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523059, China.
| |
Collapse
|
49
|
Photosynthetic microorganisms for the oxygenation of advanced 3D bioprinted tissues. Acta Biomater 2022:S1742-7061(22)00278-1. [PMID: 35562006 DOI: 10.1016/j.actbio.2022.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023]
Abstract
3D bioprinting technology has emerged as a tool that promises to revolutionize the biomedical field, including tissue engineering and regeneration. Despite major technological advancements, several challenges remain to be solved before 3D bioprinted tissues could be fully translated from the bench to the bedside. As oxygen plays a key role in aerobic metabolism, which allows energy production in the mitochondria; as a consequence, the lack of tissue oxygenation is one of the main limitations of current bioprinted tissues and organs. In order to improve tissue oxygenation, recent approaches have been established for a broad range of clinical applications, with some already applied using 3D bioprinting technologies. Among them, the incorporation of photosynthetic microorganisms, such as microalgae and cyanobacteria, is a promising approach that has been recently explored to generate chimerical plant-animal tissues where, upon light exposure, oxygen can be produced and released in a localized and controlled manner. This review will briefly summarize the state-of-the-art approaches to improve tissue oxygenation, as well as studies describing the use of photosynthetic microorganisms in 3D bioprinting technologies. STATEMENT OF SIGNIFICANCE: 3D bioprinting technology has emerged as a tool for the generation of viable and functional tissues for direct in vitro and in vivo applications, including disease modeling, drug discovery and regenerative medicine. Despite the latest advancements in this field, suboptimal oxygen delivery to cells before, during and after the bioprinting process limits their viability within 3D bioprinted tissues. This review article first highlights state-of-the-art approaches used to improve oxygen delivery in bioengineered tissues to overcome this challenge. Then, it focuses on the emerging roles played by photosynthetic organisms as novel biomaterials for bioink generation. Finally, it provides considerations around current challenges and novel potential opportunities for their use in bioinks, by comparing latest published studies using algae for 3D bioprinting.
Collapse
|
50
|
Chen H, Timashev P, Zhang Y, Xue X, Liang XJ. Nanotechnology-based combinatorial phototherapy for enhanced cancer treatment. RSC Adv 2022; 12:9725-9737. [PMID: 35424935 PMCID: PMC8977843 DOI: 10.1039/d1ra09067d] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/19/2022] [Indexed: 12/15/2022] Open
Abstract
Nanotechnology-based phototherapy has attracted enormous attention to cancer treatment owning to its non-invasiveness, high controllability and accuracy. Given the fast development of anti-tumor strategies, we summarize various examples of multifunctional nanosystems to highlight the recent advances in nanotechnology-based combinatorial phototherapy towards improving cancer treatment. The limitations of the monotherapeutic approach and the superiority of the photo-involved combinatorial strategies are discussed in each part. The future breakthroughs and clinical perspectives of combinatorial phototherapy are also outlooked. Our perspectives may inspire researchers to develop more effective phototherapy-based cancer-treating approaches.
Collapse
Affiliation(s)
- Han Chen
- School of Pharmacy, Pharm-X Center, Shanghai Jiao Tong Univeristy Shanghai 200240 China
| | - Peter Timashev
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University Moscow 119991 Russia
| | - Yuanyuan Zhang
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University Moscow 119991 Russia
| | - Xiangdong Xue
- School of Pharmacy, Pharm-X Center, Shanghai Jiao Tong Univeristy Shanghai 200240 China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China Beijing 100190 China
| |
Collapse
|