1
|
Kamble NS, Thomas S, Madaan T, Ehsani N, Sange S, Tucker K, Muhumure A, Kunkler S, Kotagiri N. Engineered bacteria as an orally administered anti-viral treatment and immunization system. Gut Microbes 2025; 17:2500056. [PMID: 40340796 PMCID: PMC12064065 DOI: 10.1080/19490976.2025.2500056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/05/2025] [Accepted: 04/24/2025] [Indexed: 05/10/2025] Open
Abstract
The emergence of new viral pathogens necessitates innovative antiviral therapies and vaccines. Traditional approaches, such as monoclonal antibodies and vaccines, are often hindered by resistance, limited effectiveness, and high costs. Here, we develop an engineered probiotic-based antiviral platform using Escherichia coli Nissle 1917 (EcN), capable of providing both mucosal and systemic immunity via oral administration. EcN was engineered to display anti-spike nanobodies or express the Spike-Receptor Binding Domain on its surface. Our findings reveal that EcN with nanobodies effectively inhibits the interaction between spike protein-expressing pseudoviruses and the ACE2 receptor. Furthermore, we observed the translocation of nanobodies to distant organs, facilitated by outer membrane vesicles (OMVs). The oral administration of EcN expressing spike proteins induced a robust immune response characterized by the production of both IgG and IgA, antibodies that blocked the pseudovirus-ACE2 interaction. While SARS-CoV-2 served as a model, this versatile probiotic platform holds potential for developing customizable biotherapeutics against a wide range of emerging pathogens such as influenza virus or respiratory syncytial virus (RSV) by engineering EcN to express viral surface protein or neutralizing nanobodies demonstrating its versatility as a next-generation mucosal vaccine strategy.
Collapse
Affiliation(s)
- Nitin S. Kamble
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Shindu Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Tushar Madaan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Nadia Ehsani
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Saqib Sange
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Kiersten Tucker
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Alexis Muhumure
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Sarah Kunkler
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
2
|
Almonte AA, Thomas S, Zitvogel L. Microbiota-centered interventions to boost immune checkpoint blockade therapies. J Exp Med 2025; 222:e20250378. [PMID: 40261296 PMCID: PMC12013646 DOI: 10.1084/jem.20250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint blockade therapies have markedly advanced cancer treatment by invigorating antitumor immunity and extending patient survival. However, therapeutic resistance and immune-related toxicities remain major concerns. Emerging evidence indicates that microbial dysbiosis diminishes therapeutic response rates, while a diverse gut ecology and key beneficial taxa correlate with improved treatment outcomes. Therefore, there is a growing understanding that manipulating the gut microbiota could boost therapy efficacy. This review examines burgeoning methods that target the gut microbiome to optimize therapy and innovative diagnostic tools to detect dysbiosis, and highlights challenges that remain to be addressed in the field.
Collapse
Affiliation(s)
- Andrew A. Almonte
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Simon Thomas
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| |
Collapse
|
3
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Das M, Kiruthiga C, Shafreen RB, Nachammai K, Selvaraj C, Langeswaran K. Harnessing the human microbiome and its impact on immuno-oncology and nanotechnology for next-generation cancer therapies. Eur J Pharmacol 2025; 996:177436. [PMID: 40023356 DOI: 10.1016/j.ejphar.2025.177436] [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: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The integration of microbiome research and nanotechnology represents a significant advancement in immuno-oncology, potentially improving the effectiveness of cancer immunotherapies. Recent studies highlight the influential role of the human microbiome in modulating immune responses, presenting new opportunities to enhance immune checkpoint inhibitors (ICIs) and other cancer therapies. Nanotechnology offers precise drug delivery and immune modulation capabilities, minimizing off-target effects while maximizing therapeutic outcomes. This review consolidates current knowledge on the interactions between the microbiome and the immune system, emphasizing the microbiome's impact on ICIs, and explores the incorporation of nanotechnology in cancer treatment strategies. Additionally, it provides a forward-looking perspective on the synergistic potential of microbiome modulation and nanotechnology to overcome existing challenges in immuno-oncology. This integrated approach may enhance the personalization and effectiveness of next-generation cancer treatments, paving the way for transformative patient care.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | | | - R Beema Shafreen
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | - Kathiresan Nachammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD Lab, Department of Microbiology, Dr. D. Y. Patil Medical College Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to Be University), Pimpri, Pune, 411018, India.
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India; Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
5
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
6
|
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] [MESH Headings] [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
|
7
|
Wang Y, Fan Y, Zhang X, Liu J, Sun D, Li L, Bai G, Liu X, Kang J, Zhang Y, Wang H. In situ production and precise release of bioactive GM-CSF and siRNA by engineered bacteria for macrophage reprogramming in cancer immunotherapy. Biomaterials 2025; 317:123037. [PMID: 39729775 DOI: 10.1016/j.biomaterials.2024.123037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/01/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
In the immunosuppressive tumor microenvironment (TME), tumor-associated macrophages (TAMs) predominantly exhibit an immunosuppressive M2 phenotype, which facilitates tumor proliferation and metastasis. Although current strategies aimed at reprogramming TAMs hold promise, their sustainability and effectiveness are limited due to repeated injections. Herein, a bacterial therapy platform containing two engineered strains was developed. One strain was engineered to produce and secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) to promote M2-like TAMs repolarization to M1-like TAMs, while the other strain was designed to secrete small interfering RNA (siRNA) targeting signal regulatory protein α (SIRPα). The two strains can continuously and efficiently produce bioactive therapeutic agents in situ, exerting a sustained and synergistic therapeutic effect in TAMs to inhibit tumor growth. To enhance treatment efficacy, optogenetic strategy was implemented to effectively control the production of GM-CSF, and outer membrane vesicles (OMVs) produced by engineered bacteria were utilized to protect the siRNA from degradation in the external environment. The experimental results indicated that the bacterial therapy platform could continuously produce and release bioactive GM-CSF and SIRPα siRNA, exhibiting significant therapeutic activity. In vivo experiments further demonstrated that this platform showed more sustained and stable therapeutic effects compared to conventional drug therapies. Additionally, the combination of these two engineered strains yielded the highest ratio of M1/M2 TAMs (0.80) and the lowest ratio of F4/80+SIRPα+TAMs (3.46 %) than single strain therapy. Our study expanded the potential of engineered bacteria as pharmaceutical factories for in vivo therapeutic applications.
Collapse
Affiliation(s)
- Yaxin Wang
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Yali Fan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xinyu Zhang
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Jing Liu
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Dawei Sun
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Lianyue Li
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Guijie Bai
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Xinyu Liu
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Jun Kang
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| | - Yingying Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hanjie Wang
- School of Life Sciences, Faculty of Medicine, Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
8
|
Kong L, Hu X, Xia D, Wu J, Zhao Y, Guo H, Zhang S, Qin C, Wang Y, Li L, Su Z, Zhu C, Xu S. Janus PEGylated CuS-engineered Lactobacillus casei combats biofilm infections via metabolic interference and innate immunomodulation. Biomaterials 2025; 317:123060. [PMID: 39736219 DOI: 10.1016/j.biomaterials.2024.123060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/25/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025]
Abstract
Bacterial implant-associated infections predominantly contribute to the failure of prosthesis implantation. The local biofilm microenvironment (BME), characterized by its hyperacidic condition and high hydrogen peroxide (H2O2) level, inhibits the host's immune response, thereby facilitating recurrent infections. Here, a Janus PEGylated CuS nanoparticle (CuPen) armed engineered Lactobacillus casei (L. casei) denoted as LC@CuPen, is proposed to interfere with bacterial metabolism and arouse macrophage antibiofilm function. Once LC@CuPen reached the BME, NIR irradiation-activated mild heat damages L. casei and biofilm structure. Meanwhile, the BME-responsive LC@CuPen can catalyze local H2O2 to produce toxic •OH, whereas in normal tissues, the effect of •OH production is greatly reduced due to the higher pH and lower H2O2 concentration. The released bacteriocin from damaged L. casei can destroy the bacterial membrane to enhance the penetration of •OH into damaged biofilm. Excessive •OH interferes with normal bacterial metabolism, resulting in reduced resistance of bacteria to heat stress. Finally, under the action of mild heat treatment, the bacterial biofilm lysed and died. Furthermore, the pathogen-associated molecular patterns (PAMPs) in LC@CuPen can induce M1 polarization of macrophages through NF-κB pathway and promote the release of inflammatory factors. Inflammatory factors enhance the migration of macrophages to the site of infection and phagocytose bacteria, thereby inhibiting the recurrence of infection. Generally, this engineered L. casei program presents a novel perspective for the treatment of bacterial implant-associated infections and serves as a valuable reference for future clinical applications of engineered probiotics.
Collapse
Affiliation(s)
- Lingtong Kong
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Xianli Hu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Demeng Xia
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jianghong Wu
- Department of Microbiology, College of Basic Medical Science, Department of Emergency, The Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
| | - Yangpeng Zhao
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Hua Guo
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Song Zhang
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Chun Qin
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Yanjun Wang
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Lei Li
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Zheng Su
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Chen Zhu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shuogui Xu
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
9
|
Zhang JT, Lezia A, Emmanuele P, Wu M, Olson EC, Somani A, Feist AM, Hasty J. Host Evolution Improves Genetic Circuit Function in Complex Growth Environments. ACS Synth Biol 2025. [PMID: 40391643 DOI: 10.1021/acssynbio.5c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
The systematic design of genetic circuits with predictable behaviors in complex environments remains a significant challenge. Here, we engineered a population control circuit and used a combination of evolutionary and rational engineering approaches to enhance Escherichia coli for robust genetic circuit behavior in nontraditional growth environments. We utilized adaptive laboratory evolution (ALE) on E. coli MG1655 in minimal media with a sole carbon source and saw improved dynamics of the circuit after host evolution. Additionally, we applied ALE to E. coli Nissle, a probiotic strain, in a more complex medium environment with added reactive oxygen species (ROS) stress. In combination with directed mutagenesis and high-throughput microfluidic screening, we observed restored circuit function and improved tolerance of the circuit components. These findings serve as a framework for the optimization of relevant bacterial host strains for improved growth and gene circuit performance in complex environments.
Collapse
Affiliation(s)
- Joanna T Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Biodynamics Laboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Andrew Lezia
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Biodynamics Laboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Philip Emmanuele
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Biodynamics Laboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Muyao Wu
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Elina C Olson
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Aayush Somani
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Biodynamics Laboratory, University of California San Diego, La Jolla, California 92093, United States
| | - Adam M Feist
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Jeff Hasty
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Biodynamics Laboratory, University of California San Diego, La Jolla, California 92093, United States
- Synthetic Biology Institute, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
10
|
Fields BD, Pascal DG, Rando OK, Huddleston ME, Ingram K, Hopton R, Grogg MW, Nelson MT, Voigt CA. Design of a Continuous GAA-Producing Probiotic as a Potential Mitigator of the Effects of Sleep Deprivation. ACS Synth Biol 2025. [PMID: 40378286 DOI: 10.1021/acssynbio.4c00690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Creatine is a popular athletic supplement that has also been shown to improve cognitive performance upon sleep deprivation. However, it is rapidly cleared from the gastrointestinal tract a few hours after consumption. Toward providing a persistent creatine dose, we engineered the human probiotic Escherichia coli Nissle (EcN) to produce guanidinoacetic acid (GAA), which is converted to creatine in the liver. We find GAA-producing enzymes present in the human microbiome and compare their activities to known enzymes. Three copies of arginine:glycine amidinotransferase (AGAT) from Actinokineospora terrae are expressed from the genome, and native gcvP, argR, and argA are edited or deleted to improve substrate availability without negatively impacting cell viability. A standard EcN dose (1012 cells) produces 41 ± 7 mg GAA per hour under laboratory conditions. This work demonstrates that a probiotic bacterium can be engineered to produce sustained GAA titers known to impact cognitive performance.
Collapse
Affiliation(s)
- Brandon D Fields
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Daniel G Pascal
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Olivia K Rando
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Mary E Huddleston
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Katherine Ingram
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Rachel Hopton
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Matthew W Grogg
- United States Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - M Tyler Nelson
- United States Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
11
|
Nama ASA, Sandeepa GM, Buddolla V, Mastan A. Advances in understanding therapeutic mechanisms of probiotics in cancer management, with special emphasis on breast cancer: A comprehensive review. Eur J Pharmacol 2025; 995:177410. [PMID: 39986595 DOI: 10.1016/j.ejphar.2025.177410] [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: 11/27/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
The increasing global prevalence of cancer, particularly breast cancer, necessitates the development of innovative therapeutic strategies. Probiotics, proficient in promoting gut health, have emerged as promising candidates for cancer treatment due to their immunomodulatory and potential anticancer properties. This review focuses on the therapeutic mechanisms of probiotics in breast cancer, examining their anticancer efficacy through metabolic, immune, and molecular mechanisms. Probiotics enhance cancer therapies, minimize side effects, and offer new adjuvant approaches in oncology. Recent advancements discussed in the review include the utilization of probiotics as oncolytic gene expression systems and drug delivery vectors, as well as personalized probiotic interventions aimed at optimizing cancer therapy. Clinical studies are critically evaluated, highlighting both the outcomes and limitations of probiotic use in cancer patients, particularly those suffering from breast cancer. Additionally, the review explores factors influencing anticancer effects of probiotics, focusing on their role in modulating the tumor microenvironment. Challenges in translating preclinical findings to clinical practice are discussed, along with future research directions, focusing on the relationship between probiotics, the microbiome, and cancer treatment. Ultimately, this review advocates for further investigation into the therapeutic potential of probiotics in breast cancer, aiming to harness their benefits in oncology.
Collapse
Affiliation(s)
- A S Angel Nama
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, 524320, India
| | - G Mary Sandeepa
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, 524320, India.
| | - Viswanath Buddolla
- Dr.Buddolla's Institute of Life Sciences (A unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India
| | - Anthati Mastan
- Dr.Buddolla's Institute of Life Sciences (A unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India.
| |
Collapse
|
12
|
Dang S, Zhang X, Zhang Y, Zhang H. New thoughts on the intestinal microbiome-B cell-IgA axis and therapies in IgA nephropathy. Autoimmun Rev 2025; 24:103835. [PMID: 40360014 DOI: 10.1016/j.autrev.2025.103835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
IgA nephropathy (IgAN), as the most common chronic glomerulonephritis worldwide, is often triggered by mucosal infections and follows a chronic progression, with the majority of patients ultimately progressing to end-stage renal disease (ESRD) during their lifetimes. Since the mystery of its complete pathogenesis has not been fully solved, the resulting lack of effective early diagnosis and treatment greatly affects the prognosis of patients. Given the well-defined pathological feature of IgA deposition in the mesangial region, the source and role of pathogenic IgA has been focused on. Starting from the microbiology and immunity of the gut, we systematically review both the physiological and the pathological process of microbiome-B cell-IgA axis, from microbial-induced IgA production to the role of IgA in the intestinal immune milieu, and ultimately end up with the various aspects of microbiome-B cell-IgA axis in the pathogenesis of IgAN as well as the corresponding therapeutic initiatives available. Our retrospective review helps researchers to systematically understand the complex role between intestinal flora dysbiosis and pathogenic IgA in IgAN. This understanding provides a foundation for in-depth explorations to uncover more detailed pathogenic mechanisms and to develop more precise and effective diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Shaoqing Dang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangyu Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yuemiao Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
13
|
Li Y, Wang X, Ye F, Hong X, Chen Y, Huang J, Liu J, Huang X, Liang L, Guo Y, Shi F, Zhu K, Lin L, Huang W. Acid-responsive engineered bacteria with aberrant In-Situ anti-PD-1 expression for post-ablation immunotherapy of hepatocellular carcinoma. Biomed Pharmacother 2025; 186:118046. [PMID: 40209305 DOI: 10.1016/j.biopha.2025.118046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Local thermal ablation (TA) can not only reduce the tumor burden of hepatocellular carcinoma (HCC) but also stimulate the host anti-tumor immune response, offering a promising avenue for combination with immune checkpoint blockade (ICB). However, tumor recurrence and ICB resistance are associated with residual tumor masses caused by incomplete TA treatment. Thus, adjuvant therapy that can accurately eliminate residual HCC tumors post-TA is expected to improve prognosis. Bacteria-mediated tumor therapy has showed promising potential for tumor-targeting ability and in situ therapeutic proteins expression in the tumor. Here, we presented a kind of nonpathogenic engineered bacteria (named PD-1@EcM) for the potent tumor-targeting and acidic-controlled production of fusion protein comprising a mouse-derived anti-PD-1 single-chain variable fragment (scFv). A single injection of this engineered bacteria demonstrated a significantly tumor inhibition and extended survival in advanced murine primary and metastatic post-TA treatment HCC model. We observed that this engineered bacteria elicited an enhanced antitumour immune response resulting in an extensive priming of activated CD8+ T cells and polarization of tumor-associated macrophage from M2 phenotype to M1 phenotype. Taken together, this work provides a novel strategy to address major challenges in TA therapy and expand the current applications of bacteria-based platforms for precision therapy.
Collapse
Affiliation(s)
- Yue Li
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Xiaobin Wang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Feilong Ye
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Xiaoyang Hong
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Ye Chen
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Jiabai Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Jianxin Liu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Xinkun Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Licong Liang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Yongjian Guo
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China
| | - Feng Shi
- Department of Interventional Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern medical university, Guangzhou, China
| | - Kangshun Zhu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China.
| | - Liteng Lin
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China.
| | - Wensou Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital, Guangzhou Medical University, Changgangdong Road, Haizhu District, Guangzhou, Guangdong Province 510261, China.
| |
Collapse
|
14
|
Shi C, Su LY, Xu W, Wang S, Cao Y, Yao M, Wang X, Liu F, Nie G, Zhou H. Smartphone-Manipulated Mild-Temperature Photothermal Control of the TfR-LYTAC Platform for Tumor Treatment. NANO LETTERS 2025; 25:6864-6871. [PMID: 40077868 DOI: 10.1021/acs.nanolett.4c06279] [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: 03/14/2025]
Abstract
A lysosome-targeting chimera (LYTAC) offers a novel strategy for degrading extracellular proteins previously considered to be undruggable by hijacking the lysosomal degradation system. However, clinical use of LYTAC has been limited due to the potential for uncontrolled degradation and systemic toxicity. Based on our previously developed genetically encoded TfR-LYTAC, here, we introduce a photothermal-inducible switch in the engineered bacterium to enable spatiotemporal expression of TfR-LYTAC. We also developed precise photothermal systems to control LYTAC expression via cellphone manipulation to avoid burns of normal tissues, especially the skin, caused by intense laser light. The engineered bacterium exhibited significant tumor-suppressing effects upon exposure to the phone-manipulated laser in both subcutaneously transplanted melanoma and colon tumor models with no skin damage to the mice. Our approach enables LYTAC precision medicine and provides a generalizable platform for developing light-driven LYTAC. Our study provides a platform for outpatient or home treatment and is a further step for telemedicine.
Collapse
Affiliation(s)
- Chunqin Shi
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650201, China
- Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Ling-Yan Su
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650201, China
- Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Wen Xu
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650201, China
| | - Shuangping Wang
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650201, China
| | - Yu Cao
- Department of Cardiovascular Surgery, The First People Hospital of Yunnan Province, Xishan District, No. 157 Jinbi Road, Kunming 650032, China
| | - Mengyu Yao
- Department of Cardiovascular Surgery, The First People Hospital of Yunnan Province, Xishan District, No. 157 Jinbi Road, Kunming 650032, China
| | - Xiao Wang
- State Key Laboratory of Mechanical Transmission, Chongqing University, Chongqing 40044, China
| | - Fei Liu
- State Key Laboratory of Mechanical Transmission, Chongqing University, Chongqing 40044, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hejiang Zhou
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650201, China
- Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
15
|
Li YR, Shen X, Zhu Y, Lyu Z, Yang L. The microbiota shapes the life trajectory of mucosal-associated invariant T cells. Trends Microbiol 2025:S0966-842X(25)00107-6. [PMID: 40280795 DOI: 10.1016/j.tim.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells predominantly located in barrier tissues such as the lung, liver, skin, and colon. These cells recognize metabolites derived from the riboflavin biosynthetic pathway, which can rapidly traverse epithelial barriers and be presented during MAIT cell differentiation in the thymus and maturation in peripheral tissues. Furthermore, microbial metabolites significantly influence MAIT cell functions in various conditions, including cancer. This review summarizes how the microbiota shapes the life trajectory of MAIT cells and their antitumor reactivity. Additionally, we discuss the therapeutic implications of manipulating the microbiota as a 'bug-drug' strategy to enhance MAIT cell antitumor immunity, particularly in mucosal cancers, while emphasizing challenges and future directions for integrating microbiota considerations into MAIT cell-based therapies.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xinyuan Shen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA; Goodman-Luskin Microbiome Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
16
|
Huang R, Kravchik V, Zaatry R, Habib M, Geva-Zatorsky N, Daniel R. Engineering coupled consortia-based biosensors for diagnostic. Nat Commun 2025; 16:3761. [PMID: 40263365 PMCID: PMC12015303 DOI: 10.1038/s41467-025-58996-9] [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: 03/04/2024] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Synthetic multicellular systems have great potential for performing complex tasks, including multi-signal detection and computation through cell-to-cell communication. However, engineering these systems is challenging, requiring precise control over the cell concentrations of distinct members and coordination of their activity. Here, we develop a bacterial consortia-based biosensor for Heme and Lactate, wherein members are coupled through a global shared quorum-sensing signal that simultaneously controls the activity of the diverse biosensing strains. The multicellular system incorporates a gene circuit that computes the minimum between each biosensor's activity and the shared signal. We evaluate three consortia configurations: one where the shared signal is externally supplied, another directly produced via an inducible gene circuit, and a third generated through an incoherent feedforward loop (IFFL) gene circuit. Among these configurations, the IFFL system, which maintains the shared signal at low and stable levels over an extended period, demonstrates improved performance and robustness against perturbations in cell populations. Finally, we examine these coupled consortia to monitor Lactate and Heme in humanized fecal samples for diagnostics.
Collapse
Affiliation(s)
- Rongying Huang
- Department of Biotechnology Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Valeriia Kravchik
- Department of Biomedical Engineering Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Rawan Zaatry
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center (RTICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, 3525422, Haifa, Israel
| | - Mouna Habib
- Department of Biomedical Engineering Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Naama Geva-Zatorsky
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center (RTICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, 3525422, Haifa, Israel
- CIFAR, MaRS Centre, West Tower 661 University Avenue, Suite 505, Toronto, ON, M5G 1M1, Canada
| | - Ramez Daniel
- Department of Biomedical Engineering Technion-Israel Institute of Technology, Technion City, Haifa, Israel.
| |
Collapse
|
17
|
Li M, Kuang C, Guo Z, Du M, Chen Z. Research progress on ultrasound in bacteria-mediated tumor treatment. Crit Rev Microbiol 2025:1-12. [PMID: 40243567 DOI: 10.1080/1040841x.2025.2489476] [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: 10/12/2024] [Revised: 03/22/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Bacteria-mediated tumor treatment (BMTT) has recently garnered significant attention as a promising avenue in tumor treatment. Despite the application of various strains in animal models and clinical trials, the effectiveness of BMTT has been hindered by its toxicity and inefficiency. In recent years, it has been explored that applying the biological effects of ultrasound could further improve the precision and effectiveness of BMTT. This review briefly introduces the challenges of BMTT and summarizes how the biological effects of ultrasound improve the efficacy and safety of BMTT in strategies involving genetic engineering, visualization and targeted delivery. The potential application and limitations of ultrasound in advancing BMTT controllable strategies are also discussed.
Collapse
Affiliation(s)
- Mingjie Li
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute for Future Sciences, University of South China, Changsha, China
| | - Chenke Kuang
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Zhili Guo
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute for Future Sciences, University of South China, Changsha, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Institute for Future Sciences, University of South China, Changsha, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Institute for Future Sciences, University of South China, Changsha, China
- Department of Medical Imaging, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
18
|
Jin M, Liu S, Zhan M, Huang JD. Engineered Genetic Circuits Activated by Bezafibrate Improve ESC-Based TAA Cancer Vaccine Efficacy and PD-L1 Nanobody Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500272. [PMID: 40245119 DOI: 10.1002/advs.202500272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/01/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy targeting tumor antigens and immune checkpoint inhibitors has garnered significant attention in cancer treatment. Synthetic gene circuits are developed, encoded in plasmids, which regulate the expression of tumor antigens shared with embryonic stem cells (ESCs) and PD-L1 nanobody (PD-L1 nb) in response to bezafibrate stimulation. This approach significantly minimizes side effects and improved therapeutic efficacy. The transcriptional switches leverage the interaction between the bezafibrate-responsive transcriptional activator PPARγ and RXRα, which are fused with the VPR/VP64/p65 activation domains (AD) and the Gal4 DNA-binding domain (DBD), respectively. These synthetic constructs are validated and their ability to modulate gene expression upon bezafibrate treatment are confirmed. Notably, the gene expression is precise and tunable in response to bezafibrate administration. HEK293T cells or ESCs are employed to deliver this gene circuit, or the plasmids containing the circuit into the tumor are directly injected. Administration of bezafibrate reduces tumor growth, increases specific CD8+ T cells, and mitigates CD8+ T cell exhaustion, underscoring the feasibility and effectiveness of the approach. ESC-based and intratumoral delivery of the synthetic gene circuits and cargo genes, particularly PD-L1 nb, significantly inhibit tumor growth. PD-L1 nb effectively blocks PD-L1 expression both in vitro and in vivo, as confirmed by using a mutant PD-L1 nb sequence.
Collapse
Affiliation(s)
- Meiling Jin
- Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Shuzhen Liu
- Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Mingshuo Zhan
- Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jian-Dong Huang
- Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, P. R. China
- Department of Clinical Oncology, Shenzhen Key Laboratory for cancer metastasis and personalized therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, P. R. China
- Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, 518057, P. R. China
| |
Collapse
|
19
|
Ahmadishoar S, Mones Saeed S, Salih Mahdi M, Mohammed Taher W, Alwan M, Jasem Jawad M, Khdyair Hamad A, Gandomkar H. The potential use of bacteria and their derivatives as delivery systems for nanoparticles in the treatment of cancer. J Drug Target 2025:1-34. [PMID: 40186857 DOI: 10.1080/1061186x.2025.2489979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Nanomaterials, unique optical, magnetic, and electrical properties at the nanoscale (1-100 nm), have been engineered to improve drug capacity, bioavailability, and specificity in cancer treatment. These advancements address toxicity and lack of selectivity in conventional therapies, enabling precise targeting of cancer cells, the tumour microenvironment, and the immune system. Among emerging approaches, bacterial treatment shows promise due to its natural ability to target cancer and its diverse therapeutic mechanisms, which nanotechnology can further enhance. Bacteria-based drug delivery systems leverage bacteria's adaptability and survival strategies within the human body. Bacterial derivatives, such as bacterial ghosts (BGs), bacterial extracellular vesicles (BEVs), and dietary toxins, are recognised as effective biological nanomaterials capable of carrying nanoparticles (NPs). These systems have attracted increasing attention for their potential in targeted NP delivery for cancer treatment. This study explores the use of various bacteria and their byproducts as NP delivery vehicles, highlighting their potential in treating different types of cancer. By combining the strengths of nanotechnology and bacterial therapy, these innovative approaches aim to revolutionise cancer treatment with improved precision and efficacy.
Collapse
Affiliation(s)
- Shiva Ahmadishoar
- Department of Microbiology, Male.C., Islamic Azad University, Malekan, Iran
| | - Samaa Mones Saeed
- Dental Prosthetics Techniques Department, Health and Medical Techniques College/AlNoor University, Mosul, Iraq
| | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | | - Hossein Gandomkar
- Department of Surgical Oncology, Tehran University of Medical Medicine, Tehran, Iran
| |
Collapse
|
20
|
Liu H, Shen S, Xu Q, Wang Y, Qi K, Lu B, Tang B, Wu M, Gan F. Noncanonical amino acids as prophage inducers for protein regulation in bacteria-based delivery systems. mBio 2025; 16:e0398824. [PMID: 40084898 PMCID: PMC11980383 DOI: 10.1128/mbio.03988-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Genetically engineered bacteria represent a promising drug delivery tool for disease treatment. The development of new strategies for specific and independent protein regulation is necessary, especially for combination protein drug therapy. Using the well-studied Escherichia coli phage λ as a model system, we applied noncanonical amino acids (ncAAs) as novel inducers for protein regulation in a bacteria-based delivery system. Screening the permissive sites of the Cro protein revealed that incorporation of AlocK at the K8 site with the MbPylRS-349F/tRNAPyl system produced a functional Cro-K8AlocK variant. Using an engineered λ lysogen expressing the MbPylRS-349F/tRNAPyl pair, Cro-8X, and the reporter mNeonGreen, in vitro and in vivo experiments showed that AlocK led to bacterial lysis through prophage activation and the release of mNeonGreen. If mNeonGreen was integrated into the λ prophage genome, λ phages released due to AlocK induction delivered the reporter gene into the recipient E. coli strain, enabling mNeonGreen expression. Furthermore, insertion of pIF at the F14 site with the AfpIFRS/tRNATyr pair produced a functional Cro-F14pIF variant. Importantly, AfpIFRS/tRNATyr and MbPylRS-349F/tRNAPyl pairs were confirmed to be mutually orthogonal. In a mixture of two engineered λ lysogens expressing different aaRS/tRNAs, Cro-ncAAs, and reporter proteins, AlocK and pIF independently induced bacterial lysis and activated the expression of mNeonGreen and mCherry in the recipient E. coli strain. Collectively, the proposed bacteria-based delivery system provides two options for protein delivery and enables independent regulation of multiple proteins with ncAAs, offering a novel approach for in situ protein regulation and combination therapy. IMPORTANCE The use of genetically engineered bacteria as drug delivery vectors has attracted more and more attention in recent years. A key issue with bacteria-based delivery systems is how to regulate multiple protein drugs. Based on genetic code expansion technology, we developed a new strategy of using ncAAs as small molecular inducers for in situ protein regulation and engineered λ phage lysogen into a bacteria-based delivery system that can function in two delivery modes. Furthermore, this strategy enables independent regulation of multiple proteins by different ncAAs, offering important implications for combination therapy. This approach requires minimal genetic engineering efforts, and similar strategies can be applied to engineer other prophage-bacteria systems or study phage biology. This work expands the therapeutic applications of ncAAs and lysogenic phages.
Collapse
Affiliation(s)
- Hongfang Liu
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Sijia Shen
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Qi Xu
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yuyang Wang
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Kejing Qi
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Bowen Lu
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Bing Tang
- Hubei Key Laboratory of Cell Homeostasis, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Min Wu
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Fei Gan
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Raeisi H, Leeflang J, Hasan S, Woods SL. Bioengineered Probiotics for Clostridioides difficile Infection: An Overview of the Challenges and Potential for This New Treatment Approach. Probiotics Antimicrob Proteins 2025; 17:763-780. [PMID: 39531149 DOI: 10.1007/s12602-024-10398-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The rapid increase in microbial antibiotic resistance in Clostridioides difficile (C. difficile) strains and the formation of hypervirulent strains have been associated with a global increase in the incidence of C. difficile infection (CDI) and subsequently, an increase in the rate of recurrence. These consequences have led to an urgent need to develop new and promising alternative strategies to control this pathogen. Engineered probiotics are exciting new bacterial strains produced by editing the genome of the original probiotics. Recently, engineered probiotics have been used to develop delivery vehicles for vaccines, diagnostics, and therapeutics. Recent studies have demonstrated engineered probiotics may potentially be an effective approach to control or treat CDI. This review provides a brief overview of the considerations for engineered probiotics for medicinal use, with a focus on recent preclinical research using engineered probiotics to prevent or treat CDI. We also address the challenges faced in the production of engineered strains and how they may be overcome in the application of these agents to meet patient needs in the future.
Collapse
Affiliation(s)
- Hamideh Raeisi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Julia Leeflang
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Sadia Hasan
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Susan L Woods
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| |
Collapse
|
22
|
Qiao L, Niu L, Wang Z, Deng Z, Di D, Ma X, Zhou Y, Kong D, Wang Q, Yin J, Jin L, Sun J, Feng B, Lu W, Cai F, Guan N, Ye H. Engineered bacteria for near-infrared light-inducible expression of cancer therapeutics. NATURE CANCER 2025; 6:612-628. [PMID: 40097656 DOI: 10.1038/s43018-025-00932-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/14/2025] [Indexed: 03/19/2025]
Abstract
Bacteria-based therapies hold great promise for cancer treatment due to their selective tumor colonization and proliferation. However, clinical application is hindered by the need for safe, precise control systems to regulate local therapeutic payload expression and release. Here we developed a near-infrared (NIR) light-mediated PadC-based photoswitch (NETMAP) system based on a chimeric phytochrome-activated diguanylyl cyclase (PadC) and a cyclic diguanylate monophosphate-dependent transcriptional activator (MrkH). The NETMAP-engineered bacteria exhibited antitumor performance in mouse tumor models with different levels of immunogenicity. Specifically, in immunogenic lymphoma tumors, NIR-induced PD-L1 and CTLA-4 nanobodies enhanced the activation of adaptive immunity. In low-immunogenic tumors-including mouse-derived colon cancer models, an orthotopic human breast cancer cell line-derived xenograft model and a colorectal cancer patient-derived xenograft model-NIR-induced azurin and cytolysin A predominantly led to tumor inhibition. Our study identifies an NIR light-mediated therapeutic platform for engineered bacteria-based therapies with customizable outputs and precise dosage control.
Collapse
Affiliation(s)
- Longliang Qiao
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lingxue Niu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Zhihao Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Zhenqiang Deng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Dai Di
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Xiaoding Ma
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Yang Zhou
- Wuhu Hospital, Health Science Center, East China Normal University, Wuhu City, China
| | - Deqiang Kong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Qilin Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Jianli Yin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Lingli Jin
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Bo Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Weiqiang Lu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Ningzi Guan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China.
| | - Haifeng Ye
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, Shanghai Academy of Natural Sciences (SANS), East China Normal University, Shanghai, China.
| |
Collapse
|
23
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [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: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
24
|
Vaaben TH, Lützhøft DO, Koulouktsis A, Dawoodi IM, Stavnsbjerg C, Kvich L, Gögenur I, Vazquez-Uribe R, Sommer MOA. Modulating tumor immunity using advanced microbiome therapeutics producing an indole metabolite. EMBO Rep 2025; 26:1688-1708. [PMID: 40055466 PMCID: PMC11977207 DOI: 10.1038/s44319-025-00386-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 04/09/2025] Open
Abstract
The gut microbiome has emerged as a key player in modulating immune responses against cancer, suggesting that microbial interventions can enhance treatment outcomes. Indole metabolites produced by probiotic bacteria activate the aryl hydrocarbon receptor (AhR), a transcription factor important for immune cell regulation. Cancer patients with high plasma concentrations of these metabolites have shown improved survival. Building on these findings, we have engineered Escherichia coli Nissle 1917 to produce the AhR agonist indole-3-acetic acid. Delivery of indole-3-acetic acid by tumor-colonizing bacteria changes the tumor microenvironment in a murine model, significantly increasing levels of CXCL9 and IFN-γ and elevating tumor-infiltrating T-cell abundance and activation. Treatment with our engineered strain inhibits tumor growth, improves survival in syngeneic tumor models, and leads to long-lasting immunity in a tumor rechallenge experiment. Further investigation indicates that this immune modulation is driven by the direct activation of AhR by indole-3-acetic acid, leading to differential cytokine expression and a shift in immune cell composition within the tumor. This study highlights the importance of microbial metabolites in immune modulation and supports exploring microbiome-based therapies in oncology.
Collapse
Affiliation(s)
- Troels Holger Vaaben
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, DK2800, Denmark
| | - Ditte Olsen Lützhøft
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, DK2800, Denmark
| | - Andreas Koulouktsis
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, DK2800, Denmark
| | - Ida Melisa Dawoodi
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Camilla Stavnsbjerg
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Lasse Kvich
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Region Zealand, 4690, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Region Zealand, 4690, Denmark
| | - Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, DK2800, Denmark
- Center for Microbiology, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | - Morten Otto Alexander Sommer
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, DK2800, Denmark.
| |
Collapse
|
25
|
Wang J, Wang J, Yu Z, Wen H, Zhao C, Zhong J, Xiao C, Li Y, Xu J, Wang J, Mao Z, Xia W. Targeting the Adenosine-Mediated Metabolic Immune Checkpoint with Engineered Probiotic for Enhanced Chemo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411813. [PMID: 39985195 PMCID: PMC12005768 DOI: 10.1002/advs.202411813] [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: 09/25/2024] [Revised: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Immunotherapy has revolutionized cancer treatment by leveraging the patient's immune system, yet its efficacy is often hampered by the immunosuppressive tumor microenvironment (TME). Adenosine, a key player in this milieu, suppresses immune cell activity via cAMP signaling. Here, an innovative strategy to remodel the TME using a genetically engineered strain of Escherichia coli Nissle 1917 that expresses adenosine deaminase on its surface under hypoxic conditions is presented. This engineered probiotic targets tumors, converts immunosuppressive adenosine to inosine, and enhances anti-tumor immune responses. In vivo, the engineered probiotic significantly improved immune cell infiltration and demonstrated synergistic effects with low-dose doxorubicin in both subcutaneous and orthotopic mouse colorectal cancer model. Furthermore, the engineered probiotic modulated the TME, promoting a shift from M2-like to M1-like macrophages and increasing effector T cell populations. These findings highlight the potential of using engineered probiotics for metabolic modulation of the TME, offering a novel approach for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Jinhui Wang
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistryIGCMESchool of ChemistrySun Yat‐Sen UniversityGuangzhou510006China
| | - Jing Wang
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistryIGCMESchool of ChemistrySun Yat‐Sen UniversityGuangzhou510006China
| | - Zhijie Yu
- Guangdong Key Laboratory of Advanced Drug DeliverySchool of Bioscience and BiopharmaceuticsGuangdong Pharmaceutical UniversityGuangzhou510006China
| | - Hongyu Wen
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistryIGCMESchool of ChemistrySun Yat‐Sen UniversityGuangzhou510006China
| | - Chensi Zhao
- State Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Jiayong Zhong
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceSun Yat‐sen UniversityGuangzhou510060China
| | - Chuanle Xiao
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceSun Yat‐sen UniversityGuangzhou510060China
| | - Yingqiu Li
- State Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Jianqiao Xu
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistryIGCMESchool of ChemistrySun Yat‐Sen UniversityGuangzhou510006China
| | - Jinquan Wang
- Guangdong Key Laboratory of Advanced Drug DeliverySchool of Bioscience and BiopharmaceuticsGuangdong Pharmaceutical UniversityGuangzhou510006China
| | - Zong‐Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistryIGCMESchool of ChemistrySun Yat‐Sen UniversityGuangzhou510006China
| | - Wei Xia
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistryIGCMESchool of ChemistrySun Yat‐Sen UniversityGuangzhou510006China
| |
Collapse
|
26
|
Vruzhaj I, Gambirasi M, Busato D, Giacomin A, Toffoli G, Safa A. Gut Microbiota-Based Immunotherapy: Engineered Escherichia coli Nissle 1917 for Oral Delivery of Glypican-1 in Pancreatic Cancer. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:633. [PMID: 40282924 PMCID: PMC12028767 DOI: 10.3390/medicina61040633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: The administration of oral vaccines offers a potential strategy for cancer immunotherapy; yet, the development of effective platforms continues to pose a difficulty. This study examines Escherichia coli Nissle 1917 (EcN) as a microbial vector for the precise delivery of Glypican-1 (GPC1), a tumor-associated antigen significantly overexpressed in pancreatic ductal adenocarcinoma (PDAC).To evaluate the effectiveness of EcN as a vector for the delivery of GPC1 and assess its potential as an oral vaccination platform for cancer immunotherapy. Materials and Methods: EcN was genetically modified to produce a GPC1-flagellin fusion protein (GPC1-FL) to augment antigen immunogenicity. The expression and stability of GPC1 were confirmed in modified PANC02 cells using Western blot and flow cytometry, indicating that GPC1 expression did not influence tumor cell growth. A mouse model was employed to test immunogenicity post-oral delivery, measuring systemic IgG, IL-10, IL-2, and IFN-γ levels to indicate immune activation. Results: Oral immunization with EcN GPC1-FL elicited a robust systemic immune response, demonstrated by markedly increased levels of IgG and IL-10. IL-2 and IFN-γ concentrations were elevated in vaccinated mice relative to controls; however, the differences lacked statistical significance. Western blot examination of fecal samples verified consistent antigen expression in the gastrointestinal tract, indicating effective bacterial colonization and antigen retention. No detrimental impacts were noted, hence substantiating the safety of this methodology. Conclusions: These findings confirm EcN as a feasible and patient-friendly oral vaccination platform for cancer immunotherapy. The effective production of GPC1 in tumor cells, along with continuous antigen delivery and immune activation, underscores the promise of this approach for PDAC and other cancers. This study promotes microbial-based antigen delivery as a scalable, non-invasive substitute for traditional vaccine platforms.
Collapse
Affiliation(s)
- Idris Vruzhaj
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Marta Gambirasi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Aurora Giacomin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Amin Safa
- Doctoral School in Pharmacological Sciences, University of Padua, 35122 Padova, Italy
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
27
|
Jiang N, Jiang M, Chen J, Mohsin A, Mu Y, Yi X, Zhuang Y, Qian J, Huang J. Living hybrid material based on probiotic with photothermal properties inhibits PD-L1 expression after tumouricidal photothermal therapy. BIOMATERIALS TRANSLATIONAL 2025; 6:73-84. [PMID: 40313568 PMCID: PMC12041808 DOI: 10.12336/biomatertransl.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/22/2024] [Accepted: 02/25/2025] [Indexed: 05/03/2025]
Abstract
Photothermal therapy is a safe and effective tumour treatment strategy due to its excellent spatiotemporal controllability. However, interferon gamma in the tumour microenvironment is upregulated after photothermal therapy, which enhances the expression of programmed cell death ligand 1 (PD-L1) in tumour cells. This further promotes immunosuppression and tumour metastasis, resulting in a poor prognosis in cancer therapy. Traditional nanodrugs often face challenges in penetrating the dense extracellular matrix of solid tumours, whereas certain probiotics possess the ability to specifically colonise the core regions of tumours. In this research, we used Escherichia coli Nissle 1917 (ECN) as a chassis cell and self-assembly polydopamine (PDA) on the ECN surface. The black PDA@ECN (notes as PE) actively colonises at the tumour site and produces a photothermal effect under 808 nm laser irradiation to kill tumour cells. To overcome the high expression of PD-L1 induced after photothermal therapy, metformin (MET) was also encapsulated in PE to form PDA@MET@ECN (notes as PME). In vivo experiments demonstrated that PME effectively inhibited the PD-L1 expression and growth of CT26 tumour cells. Overall, PME reverses the immunosuppressive tumour microenvironment and enhances the effect of photothermal/immune therapy in tumour treatment.
Collapse
Affiliation(s)
- Ning Jiang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Mingyan Jiang
- College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, China
| | - Jianshu Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuqing Mu
- Institute for biomedicine and glycomics, Griffith University School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Xiaoping Yi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jiangchao Qian
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jiaofang Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, China
| |
Collapse
|
28
|
Liu QL, Zhou H, Wang Z, Chen Y. Exploring the role of gut microbiota in colorectal liver metastasis through the gut-liver axis. Front Cell Dev Biol 2025; 13:1563184. [PMID: 40181829 PMCID: PMC11965903 DOI: 10.3389/fcell.2025.1563184] [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: 01/19/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Colorectal liver metastasis (CRLM) represents a major therapeutic challenge in colorectal cancer (CRC), with complex interactions between the gut microbiota and the liver tumor microenvironment (TME) playing a crucial role in disease progression via the gut-liver axis. The gut barrier serves as a gatekeeper, regulating microbial translocation, which influences liver colonization and metastasis. Through the gut-liver axis, the microbiota actively shapes the TME, where specific microbial species and their metabolites exert dual roles in immune modulation. The immunologically "cold" nature of the liver, combined with the influence of the gut microbiota on liver immunity, complicates effective immunotherapy. However, microbiota-targeted interventions present promising strategies to enhance immunotherapy outcomes by modulating the gut-liver axis. Overall, this review highlights the emerging evidence on the role of the gut microbiota in CRLM and provides insights into the molecular mechanisms driving the dynamic interactions within the gut-liver axis.
Collapse
Affiliation(s)
- Qiu-Luo Liu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Institute of Digestive Surgery, Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Huijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Health Management Center, General Practice Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Chen
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
29
|
Li X, Wang Y, Wang Y, Xie H, Gong R, Wu X, Chen J, Sun C, Gu Y. Anti-tumor activity of an αPD-L1-PE38 immunotoxin delivered by engineered Nissle 1917. Int J Biol Macromol 2025; 295:139537. [PMID: 39788238 DOI: 10.1016/j.ijbiomac.2025.139537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Although immune checkpoint inhibitors specifically targeting the PD-1/PD-L1 axis have exhibited remarkable clinical success, they are not uniformly effective across all patient cohorts. Immunotoxins, a novel class of cancer therapeutics, offering a promising alternative. PD-L1, which is also present in certain normal tissues, limits its suitability as an ideal target for immunotoxins. The probiotic strain of E. coli Nissle 1917 (EcN) could target and colonize to solid tumors, which positions it as a promising candidate for tumor tissue-specific delivery of anti-tumor proteins. In this study, we constructed a PD-L1-targeted immunotoxin, designated as αPD-L1-PE38, by fusing an anti-PD-L1 nanobody and a clinically validated PE38 toxin. This immunotoxin exhibited potent cytotoxic activity against tumor cells while showed slightly cytotoxic activity against normal cells. To effectively deliver the αPD-L1-PE38 to tumor tissues, we engineered the EcN strain to release the immunotoxin induced by L-arabinose. Upon induction, the immunotoxin was efficiently secreted, and exhibited robust anti-tumor activity mainly by inducing cell apoptosis both in vitro and in vivo. Furthermore, we enhanced the immunotoxin's affinity for PD-L1 by optimizing the linker between the nanobody and PE38 toxin. The engineered EcN expressing the optimized immunotoxin, achieved superior anti-tumor activity. Collectively, our study suggests that the delivery of immunotoxins through live bacteria to improve safety and efficacy is a promising option in cancer therapeutics.
Collapse
Affiliation(s)
- Xinrui Li
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yangui Wang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yanqing Wang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Huilin Xie
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Ruxin Gong
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiao Wu
- Department of Respiratory and Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, China
| | - Jin Chen
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Changning Sun
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Yuchao Gu
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
30
|
Zheng L, Wang H, Zhong X, Jia L, Shi G, Bai C, Yang R, Huang Z, Jiang Y, Wei J, Dong Z, Li J, Long Y, Dai L, Li Z, Chen C, Wang J. Reprogramming tumor microenvironment with precise photothermal therapy by calreticulin nanobody-engineered probiotics. Biomaterials 2025; 314:122809. [PMID: 39303415 DOI: 10.1016/j.biomaterials.2024.122809] [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: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024]
Abstract
Targeted therapies have revolutionized traditional cancer treatments by precisely targeting tumor cells, enhancing efficacy and safety. Despite this advancement, the proportion of cancer patients eligible for such therapies remains low due to the absence of suitable targets. Here, we investigate whether the translocation of the immunogenic cell death (ICD) marker calreticulin (CALR) from the endoplasmic reticulum (ER) to the cell surface following ICD induction can serve as a target for targeted therapies. To target CALR, a nanobody Nb215 identified from a naïve VHH phage library with high binding affinity to both human and mouse CALR was employed to engineer probiotic EcN 1917. Our results demonstrated that CALR nanobody-modified EcN-215 coupled with the photothermal dye indocyanine green (ICG) was able to exert NIR-II imaging-guide photothermal therapy (PTT). Moreover, PTT with EcN-215/ICG can reshape the tumor microenvironment by enhancing the infiltration of CD45+CD3+ T cells and CD11b+F4/80+ macrophages. Furthermore, the antitumor activity of CALR-targeted EcN-215/ICG is synergistically enhanced by blocking CD47-SIRPα axis. Collectively, our study provides a proof of concept for CALR-targeted therapy. Given that CALR translocation can be induced by various anticancer therapies across numerous tumor cell lines, CALR-targeted therapies hold promise as a novel approach for treating multiple types of cancers.
Collapse
Affiliation(s)
- Liuhai Zheng
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Huifang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaoru Zhong
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Lin Jia
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Guangwei Shi
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528399, China
| | - Chongzhi Bai
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Runwei Yang
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528399, China
| | - Zhenhui Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Yuke Jiang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Jinxi Wei
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Zhiyu Dong
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Jiexuan Li
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Ying Long
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Lingyun Dai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Zhijie Li
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China.
| | - Chunbo Chen
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Jigang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
31
|
Chen H, Li Y, Li Z, Sun Y, Gu W, Chen C, Cheng Y. Bacterial Autonomous Intelligent Microrobots for Biomedical Applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70011. [PMID: 40235203 DOI: 10.1002/wnan.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
Micro/nanorobots are being increasingly utilized as new diagnostic and therapeutic platforms in the biomedical field, enabling remote navigation to hard-to-reach tissues and the execution of various medical procedures. Although significant progress has been made in the development of biomedical micro/nanorobots, how to achieve closed-loop control of them from sensing, memory, and precise trajectory planning to feedback to carry out biomedical tasks remains a challenge. Bacteria with self-propulsion and autonomous intelligence properties are well suited to be engineered as microrobots to achieve closed-loop control for biomedical applications. By virtue of synthetic biology, bacterial microrobots possess an expanded genetic toolbox, allowing them to load input sensors to respond or remember external signals. To achieve accurate control in the complex physiological environment, the development of bacterial microrobots should be matched with the corresponding control system design. In this review, a detailed summary of the sensing and control mechanisms of bacterial microrobots is presented. The engineering and applications of bacterial microrobots in the biomedical field are highlighted. Their future directions of bacterial autonomous intelligent microrobots for precision medicine are forecasted.
Collapse
Affiliation(s)
- Haotian Chen
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingze Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhenguang Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuantai Sun
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weicheng Gu
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Cheng
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
32
|
Wang R, Li W, Cao H, Zhang L. Decoding the Tumor-Associated Microbiota: From Origins to Nanomedicine Applications in Cancer Therapy. BIOLOGY 2025; 14:243. [PMID: 40136500 PMCID: PMC11940167 DOI: 10.3390/biology14030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Growing evidence reveals that the tumor microbiome-comprising distinct microbial communities within neoplastic tissues-exerts a profound influence on cancer initiation, progression, and therapeutic response. These microbes actively reshape the tumor microenvironment (TME) through metabolite secretion, the modulation of immune pathways, and direct interactions with host cells, thereby affecting tumor biology and therapeutic outcomes. Despite substantial heterogeneity among cancer types, recent insights underscore the tumor microbiome's potential as both a diagnostic/prognostic biomarker and a targetable component for innovative treatments. In this review, we synthesize emerging knowledge on the mechanistic roles of tumor-associated microbiota in shaping the TME, with a focus on how these discoveries can guide novel therapeutic strategies. We further explore interdisciplinary advances, including the convergence of microbiomics and nanotechnology, to enhance drug delivery, circumvent resistance, and foster TME remodeling. By highlighting these cutting-edge developments, our review underscores the transformative potential of integrating tumor microbiome research into precision oncology and advancing more personalized cancer therapies.
Collapse
Affiliation(s)
- Ruiqi Wang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
| | - Weizheng Li
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
| | - Hongqian Cao
- Department of Health Inspection and Quarantine, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
33
|
Yang M, Zhong P, Wei P. Living Bacteria: A New Vehicle for Vaccine Delivery in Cancer Immunotherapy. Int J Mol Sci 2025; 26:2056. [PMID: 40076679 PMCID: PMC11900161 DOI: 10.3390/ijms26052056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer vaccines, aimed at evolving the human immune system to eliminate tumor cells, have long been explored as a method of cancer treatment with significant clinical potential. Traditional delivery systems face significant challenges in directly targeting tumor cells and delivering adequate amounts of antigen due to the hostile tumor microenvironment. Emerging evidence suggests that certain bacteria naturally home in on tumors and modulate antitumor immunity, making bacterial vectors a promising vehicle for precision cancer vaccines. Live bacterial vehicles offer several advantages, including tumor colonization, precise drug delivery, and immune stimulation, making them a compelling option for cancer immunotherapy. In this review, we explore the mechanisms of action behind living bacteria-based vaccines, recent progress in popular bacterial chassis, and strategies for specific payload delivery and biocontainment to ensure safety. These approaches will lay the foundation for developing an affordable, widely applicable cancer vaccine delivery system. This review also discusses the challenges and future opportunities in harnessing bacterial-based vaccines for enhanced therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
| | | | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (M.Y.); (P.Z.)
| |
Collapse
|
34
|
Gong X, Liu S, Xia B, Wan Y, Zhang S, Zhang B, Wang Z, Chen J, Xiao F, Liang XJ, Yang Y. Oral delivery of therapeutic proteins by engineered bacterial type zero secretion system. Nat Commun 2025; 16:1862. [PMID: 39984501 PMCID: PMC11845744 DOI: 10.1038/s41467-025-57153-6] [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: 05/21/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
Genetically engineered commensal bacteria are promising living drugs, however, their therapeutic molecules are frequently confined to their colonization sites. Herein, we report an oral protein delivery technology utilizing an engineered bacterial type zero secretion system (T0SS) via outer membrane vesicles (OMVs). We find that OMVs produced in situ by Escherichia coli Nissle 1917 (EcN) can penetrate the intact gut epithelial barrier to enter the circulation and that epithelial transcytosis involves pinocytosis and dynamin-dependent pathways. EcN is engineered to endogenously load various enzymes into OMVs, and the secreted enzyme-loaded OMVs are able to stably catalyze diverse detoxification reactions against digestive fluid and even enter the circulation. Using hyperuricemic mice and uricase delivery as a demonstration, we demonstrate that the therapeutic efficacy of our engineered EcN with a modified T0SS outperforms that with a direct protein secretion apparatus. The enzyme-loaded OMVs also effectively detoxify human serum samples, highlighting the potential for the clinical treatment of metabolic disorders.
Collapse
Affiliation(s)
- Xu Gong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Shan Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Bozhang Xia
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yichen Wan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Shuyi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, P. R. China
- Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, P. R. China
| | - Baoyan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Zehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, P. R. China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China.
| | - Yun Yang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China.
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China.
| |
Collapse
|
35
|
Zahedifard Z, Mahmoodi S, Ghasemian A. Genetically Engineered Bacteria as a Promising Therapeutic Strategy Against Cancer: A Comprehensive Review. Biotechnol Appl Biochem 2025. [PMID: 39985148 DOI: 10.1002/bab.2738] [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: 09/01/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
As a significant cause of global mortality, the cancer has also economic impacts. In the era of cancer therapy, mitigating side effects and costs and overcoming drug resistance is crucial. Microbial species can grow inside the tumor microenvironment and inhibit cancer growth through direct killing of tumor cells and immunoregulatory effects. Although microbiota or their products have demonstrated anticancer effects, the possibility of acting as pathogens and exerting side effects in certain individuals is a risk. Hence, several genetically modified/engineered bacteria (GEB) have been developed to this aim with ability of diagnosing and selective targeting and destruction of cancers. Additionally, GEB are expected to be considerably more efficient, safer, more permeable, less costly, and less invasive theranostic approaches compared to wild types. Potential GEB strains such as Escherichia coli (Nissle 1917, and MG1655), Salmonella typhimurium YB1 SL7207 (aroA gene deletion), VNP20009 (∆msbB/∆purI) and ΔppGpp (PTet and PBAD), and Listeria monocytogenes Lmat-LLO have been developed to combat cancer cells. When used in tandem with conventional treatments, GEB substantially improve the efficacy of anticancer therapy outcomes. In addition, public acceptance, optimal timing (s), duration (s), dose (s), and strains identification, interactions with other strains and the host cells, efficacy, safety and quality, and potential risks and ethical dilemmas include major challenges.
Collapse
Affiliation(s)
- Zahra Zahedifard
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
36
|
Xu H, Wang Y, Liu G, Zhu Z, Shahbazi M, Reis RL, Kundu SC, Shi X, Zu M, Xiao B. Nano-Armed Limosilactobacillus reuteri for Enhanced Photo-Immunotherapy and Microbiota Tryptophan Metabolism against Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410011. [PMID: 39739630 PMCID: PMC11831460 DOI: 10.1002/advs.202410011] [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: 08/21/2024] [Revised: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Despite being a groundbreaking approach to treating colorectal cancer (CRC), the efficacy of immunotherapy is significantly compromised by the immunosuppressive tumor microenvironment and dysbiotic intestinal microbiota. Here, leveraging the superior carrying capacity and innate immunity-stimulating property of living bacteria, a nanomedicine-engineered bacterium, LR-S-CD/CpG@LNP, with optical responsiveness, immune-stimulating activity, and the ability to regulate microbiota metabolome is developed. Immunoadjuvant (CpG) and carbon dot (CD) co-loaded plant lipid nanoparticles (CD/CpG@LNPs) are constructed and conjugated to the surface of Limosilactobacillus reuteri (LR) via reactive oxygen species (ROS)-responsive linkers. The inherent photothermal and photodynamic properties of oral CD/CpG@LNPs induce in situ cytotoxic ROS generation and immunogenic cell death of colorectal tumor cells. The generated neoantigens and the released CpG function as a potent in situ vaccine that stimulates the maturation of immature dendritic cells. The mature dendritic cells and metabolites secreted by LR subsequently facilitated the tumor infiltration of cytotoxic T lymphocytes to eradicate colorectal tumors. The further in vivo results demonstrate that the photo-immunotherapy and intestinal microbial metabolite regulation of LR-S-CD/CpG@LNPs collectively suppressed the growth of orthotopic colorectal tumors and their liver metastases, presenting a promising avenue for synergistic treatment of CRC via the oral route.
Collapse
Affiliation(s)
- Haiting Xu
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Yajun Wang
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and TechnologyChengdu610054China
| | - Ga Liu
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Zhenhua Zhu
- Department of GastroenterologyThe First Affiliated Hospital of Nanchang UniversityNanchang330006China
| | - Mohammad‐Ali Shahbazi
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenAntonius Deusinglaan 1Groningen9713 AVNetherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity of GroningenAntonius Deusinglaan 1Groningen9713 AVNetherlands
| | - Rui L. Reis
- 3Bs Research GroupI3Bs — Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, BarcoGuimarães4805‐017Portugal
- ICVS/3B's‐PT Government Associate LaboratoryBragaGuimarães4800‐058Portugal
| | - Subhas C. Kundu
- 3Bs Research GroupI3Bs — Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, BarcoGuimarães4805‐017Portugal
- ICVS/3B's‐PT Government Associate LaboratoryBragaGuimarães4800‐058Portugal
| | - Xiaoxiao Shi
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Menghang Zu
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Bo Xiao
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and TechnologyChengdu610054China
| |
Collapse
|
37
|
Mehrotra S, Kaur N, Kaur S, Matharoo K, Pandey RK. From antibodies to nanobodies: The next frontier in cancer theranostics for solid tumors. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:287-329. [PMID: 39978969 DOI: 10.1016/bs.apcsb.2024.10.014] [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: 02/22/2025]
Abstract
The field of cancer therapeutics has witnessed significant advancements over the past decades, particularly with the emergence of immunotherapy. This chapter traces the transformative journey from traditional antibody-based therapies to the innovative use of nanobodies in the treatment and diagnosis of solid tumors. Nanobodies are the smallest fragments of antibodies derived from camelid immunoglobulins and have redefined the possibilities in cancer theranostics due to their unique structural and functional properties. We provide an overview of the biochemical characteristics of nanobodies that make them particularly suitable for theranostic applications, such as their small size, high stability, enhanced infiltration into the complex tumor microenvironment (TME) and ability to bind with high affinity to epitopes that are inaccessible to conventional antibodies. Further, their ease of modification and functionalization has enabled the development of nanobody-based drug conjugates/toxins and radiolabeled compounds for precise imaging and targeted radiotherapy. We elucidate how nanobodies are being served as valuable tools for prognostic assessment, enabling clinicians to predict disease aggressiveness, monitor treatment response, and stratify patients for personalized therapeutic interventions.
Collapse
Affiliation(s)
- Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Navdeep Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sukhpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Kawaljit Matharoo
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | |
Collapse
|
38
|
Rebeck ON, Wallace MJ, Prusa J, Ning J, Evbuomwan EM, Rengarajan S, Habimana-Griffin L, Kwak S, Zahrah D, Tung J, Liao J, Mahmud B, Fishbein SRS, Ramirez Tovar ES, Mehta R, Wang B, Gorelik MG, Helmink BA, Dantas G. A yeast-based oral therapeutic delivers immune checkpoint inhibitors to reduce intestinal tumor burden. Cell Chem Biol 2025; 32:98-110.e7. [PMID: 39571582 PMCID: PMC11741927 DOI: 10.1016/j.chembiol.2024.10.013] [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: 03/15/2024] [Revised: 08/09/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Engineered probiotics are an emerging platform for in situ delivery of therapeutics to the gut. Herein, we developed an orally administered, yeast-based therapeutic delivery system to deliver next-generation immune checkpoint inhibitor (ICI) proteins directly to gastrointestinal tumors. We engineered Saccharomyces cerevisiae var. boulardii (Sb), a probiotic yeast with high genetic tractability and innate anticancer activity, to secrete "miniature" antibody variants that target programmed death ligand 1 (Sb_haPD-1). When tested in an ICI-refractory colorectal cancer (CRC) mouse model, Sb_haPD-1 significantly reduced intestinal tumor burden and resulted in significant shifts to the immune cell profile and microbiome composition. This oral therapeutic platform is modular and highly customizable, opening new avenues of targeted drug delivery that can be applied to treat a myriad of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Olivia N Rebeck
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Miranda J Wallace
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jerome Prusa
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Esse M Evbuomwan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Sunaina Rengarajan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Dermatology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis MO 63110, USA
| | - LeMoyne Habimana-Griffin
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suryang Kwak
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Zahrah
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason Tung
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James Liao
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bejan Mahmud
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erick S Ramirez Tovar
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rehan Mehta
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark G Gorelik
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beth A Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
39
|
Jin K, Yu L, Zhang Y, Niu L, Huang Y, Zhang Y, Wu Y. A Microrobotic System Based on Engineered Bacteria for Targeted Self-Driven Photodynamic Therapy. Angew Chem Int Ed Engl 2025; 64:e202414347. [PMID: 39607388 DOI: 10.1002/anie.202414347] [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: 07/29/2024] [Indexed: 11/29/2024]
Abstract
Photodynamic therapy (PDT) has been used clinically to treat superficial tumors for decades. However, its effectiveness against deep-seated tumors has been limited by the inefficient delivery of the key components -light, photosensitizer, and oxygen- required for the photochemical reactions in PDT. Here, we present a novel platform that enables the photochemical reaction to occur in a self-driven manner, eliminating the need for external delivery of these components and instead orchestrating their endogenous generation within tumors. This was achieved by genetically modifying probiotic Escherichia coli to host three modules - Lux, Hem1, and KatG - responsible for light production, photosensitizer biosynthesis, and oxygen generation, respectively. The system is self-driven, relying solely on substrates within E. coli cells and tumors. The modules exhibited prolonged activity for days within in vivo mouse models, enabling metronomic PDT that induced an immune response. This research holds promise for revolutionizing PDT and overcoming the enduring challenges encountered in its application for treating deep-seated tumors.
Collapse
Affiliation(s)
- Kai Jin
- Department of Environmental and Chemical Engineering, Shanghai University, Nanchen Rd. 333, Shanghai, China
| | - Lin Yu
- Department of Environmental and Chemical Engineering, Shanghai University, Nanchen Rd. 333, Shanghai, China
- School of Medicine, Shanghai University, Nanchen Rd. 333, Shanghai, China
- School of Medicine, Zhejiang University, Yuhangtang Rd. 333, Hangzhou, China
| | - Yue Zhang
- Department of Environmental and Chemical Engineering, Shanghai University, Nanchen Rd. 333, Shanghai, China
| | - Luqi Niu
- Department of Environmental and Chemical Engineering, Shanghai University, Nanchen Rd. 333, Shanghai, China
| | - Yi Huang
- Department of Environmental and Chemical Engineering, Shanghai University, Nanchen Rd. 333, Shanghai, China
| | - Yong Zhang
- Department of Biomedical Engineering, The City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Yihan Wu
- Department of Environmental and Chemical Engineering, Shanghai University, Nanchen Rd. 333, Shanghai, China
| |
Collapse
|
40
|
Manole S, Nguyen DH, Min JJ, Zhou S, Forbes N. Setting "cold" tumors on fire: Cancer therapy with live tumor-targeting bacteria. MED 2025; 6:100549. [PMID: 39689707 DOI: 10.1016/j.medj.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/18/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Immunotherapy with checkpoint blockade has shown remarkable efficacy in many patients with a variety of different types of cancer. However, the majority of patients with cancer have yet to benefit from this revolutionary therapy. Studies have shown that checkpoint blockade works best against immune-inflamed tumors characterized by the presence of tumor-infiltrating lymphocytes (TILs). In this review, we summarize studies using live tumor-targeting bacteria to treat cancer and describe various strategies to engineer the tumor-targeting bacteria for maximized immunoregulatory effects. We propose that tumor-localized infections by such engineered bacteria can create an immune microenvironment in favor of a more effective antitumor immunity with or without other therapies, such as immune checkpoint blockade (ICB). Finally, we will briefly outline some exemplary oncology clinical trials involving ICB plus live therapeutic bacteria, with a focus on their ability to modulate antitumor immune responses.
Collapse
Affiliation(s)
- Simin Manole
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeonnam 58128, South Korea.
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Neil Forbes
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA; Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA; Department of Microbiology, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
41
|
Yan S, Gan Y, Xu H, Piao H. Bacterial carrier-mediated drug delivery systems: a promising strategy in cancer therapy. Front Bioeng Biotechnol 2025; 12:1526612. [PMID: 39845371 PMCID: PMC11750792 DOI: 10.3389/fbioe.2024.1526612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cancer is a major killer threatening modern human health and a leading cause of death worldwide. Due to the heterogeneity and complexity of cancer, traditional treatments have limited effectiveness. To address this problem, an increasing number of researchers and medical professionals are working to develop new ways to treat cancer. Bacteria have chemotaxis that can target and colonize tumor tissue, as well as activate anti-tumor immune responses, which makes them ideal for biomedical applications. With the rapid development of nanomedicine and synthetic biology technologies, bacteria are extensively used as carriers for drug delivery to treat tumors, which holds the promise of overcoming the limitations of conventional cancer treatment regimens. This paper summarizes examples of anti-cancer drugs delivered by bacterial carriers, and their strengths and weaknesses. Further, we emphasize the promise of bacterial carrier delivery systems in clinical translation.
Collapse
Affiliation(s)
- Sizuo Yan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Yu Gan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Huizhe Xu
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
- Central Laboratory, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
42
|
Liu Z, Wang L, Wu P, Yuan L. Precision tumor treatment utilizing bacteria: principles and future perspectives. Appl Microbiol Biotechnol 2025; 109:2. [PMID: 39754636 DOI: 10.1007/s00253-024-13378-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/06/2025]
Abstract
Bacteria-based tumor therapy, which releases therapeutic payloads or remodels the tumor's immune-suppressive microenvironment and directly kills tumor cells or initiates an anti-tumor immune response, is recently recognized as a promising strategy. Bacteria could be endowed with the capacities of tumor targeting, tumor cell killing, and anti-tumor immune activating by established gene engineering. Furthermore, the integration of synthetic biology and nanomedicine into these engineered bacteria could further enhance their efficacy and controllability. This comprehensive review systematically elucidates the classification and mechanisms of bacterial gene expression induction systems, as well as strategies for constructing bacterial-nanomaterial nanobiohybrids. The review concludes by highlighting the challenges associated with quality control and regulation of bacteria-based tumor therapy while also providing insights into the future prospects of this therapeutic technology. KEY POINTS: • A comprehensive overview of the current status of research on bacteria-based tumor therapy. • The classification and mechanisms of bacterial gene expression induction systems are summarized. • The challenges and perspectives in clinical translation.
Collapse
Affiliation(s)
- Zhaoyou Liu
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Lantian Wang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Pengying Wu
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Lijun Yuan
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
43
|
Cao Y, Yan W, Yi W, Yin Q, Li Y. Bioengineered therapeutic systems for improving antitumor immunity. Natl Sci Rev 2025; 12:nwae404. [PMID: 40114728 PMCID: PMC11925021 DOI: 10.1093/nsr/nwae404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/29/2024] [Accepted: 11/08/2024] [Indexed: 03/22/2025] Open
Abstract
Immunotherapy, a monumental advancement in antitumor therapy, still yields limited clinical benefits owing to its unguaranteed efficacy and safety. Therapeutic systems derived from cellular, bacterial and viral sources possess inherent properties that are conducive to antitumor immunotherapy. However, crude biomimetic systems have restricted functionality and may produce undesired toxicity. With advances in biotechnology, various toolkits are available to add or subtract certain properties of living organisms to create flexible therapeutic platforms. This review elaborates on the creation of bioengineered systems, via gene editing, synthetic biology and surface engineering, to enhance immunotherapy. The modifying strategies of the systems are discussed, including equipment for navigation and recognition systems to improve therapeutic precision, the introduction of controllable components to control the duration and intensity of treatment, the addition of immunomodulatory components to amplify immune activation, and the removal of toxicity factors to ensure biosafety. Finally, we summarize the advantages of bioengineered immunotherapeutic systems and possible directions for their clinical translation.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenzhe Yi
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
44
|
Jin K, Huang Y, Che H, Wu Y. Engineered Bacteria for Disease Diagnosis and Treatment Using Synthetic Biology. Microb Biotechnol 2025; 18:e70080. [PMID: 39801378 PMCID: PMC11725985 DOI: 10.1111/1751-7915.70080] [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: 07/04/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025] Open
Abstract
Using synthetic biology techniques, bacteria have been engineered to serve as microrobots for diagnosing diseases and delivering treatments. These engineered bacteria can be used individually or in combination as microbial consortia. The components within these consortia complement each other, enhancing diagnostic accuracy and providing synergistic effects that improve treatment efficacy. The application of microbial therapies in cancer, intestinal diseases, and metabolic disorders underscores their significant potential. The impact of these therapies on the host's native microbiota is crucial, as engineered microbes can modulate and interact with the host's microbial environment, influencing treatment outcomes and overall health. Despite numerous advancements, challenges remain. These include ensuring the long-term survival and safety of bacteria, developing new chassis microbes and gene editing techniques for non-model strains, minimising potential toxicity, and understanding bacterial interactions with the host microbiota. This mini-review examines the current state of engineered bacteria and microbial consortia in disease diagnosis and treatment, highlighting advancements, challenges, and future directions in this promising field.
Collapse
Affiliation(s)
- Kai Jin
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yi Huang
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Hailong Che
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yihan Wu
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| |
Collapse
|
45
|
Vazquez-Uribe R, Hedin KA, Licht TR, Nieuwdorp M, Sommer MOA. Advanced microbiome therapeutics as a novel modality for oral delivery of peptides to manage metabolic diseases. Trends Endocrinol Metab 2025; 36:29-41. [PMID: 38782649 DOI: 10.1016/j.tem.2024.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
The rising prevalence of metabolic diseases calls for innovative treatments. Peptide-based drugs have transformed the management of conditions such as obesity and type 2 diabetes. Yet, challenges persist in oral delivery of these peptides. This review explores the potential of 'advanced microbiome therapeutics' (AMTs), which involve engineered microbes for delivery of peptides in situ, thereby enhancing their bioavailability. Preclinical work on AMTs has shown promise in treating animal models of metabolic diseases, including obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease. Outstanding challenges toward realizing the potential of AMTs involve improving peptide expression, ensuring predictable colonization control, enhancing stability, and managing safety and biocontainment concerns. Still, AMTs have potential for revolutionizing the treatment of metabolic diseases, potentially offering dynamic and personalized novel therapeutic approaches.
Collapse
Affiliation(s)
- Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Karl Alex Hedin
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Morten O A Sommer
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark.
| |
Collapse
|
46
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2831-x. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
47
|
Wang H, Xu F, Yao C, Dai H, Xu J, Wu B, Tian B, Shi X, Wang C. Engineering bacteria for cancer immunotherapy by inhibiting IDO activity and reprogramming CD8+ T cell response. Proc Natl Acad Sci U S A 2024; 121:e2412070121. [PMID: 39693352 DOI: 10.1073/pnas.2412070121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting indoleamine 2,3 dioxygenase (IDO) for anticancer therapy has garnered significant attention in recent years. However, current IDO inhibitors face significant challenges which limit their clinical application. Here, we genetically engineered a high tryptophan-expressing Clostridium butyricum (L-Trp CB) strain that can colonize tumors strictly following systemic administration. We revealed that butyrate produced by L-Trp CB can inhibit IDO activity, preventing tryptophan catabolism and kynurenine accumulation in tumors. In addition, the large released tryptophan by L-Trp CB can provide discrete signals that support CD8+ T cell activation and energy metabolism within the tumor microenvironment. We observed that L-Trp CB significantly restored the proportion and function of CD8+ T cells, leading to significantly delayed tumor growth in both mouse and rabbit multiple tumor models with limited side effects. We here provide a synthetic biology treatment strategy for enhanced tumor immunotherapy by inhibiting IDO activity and reprogramming CD8+ T cell response in tumors.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenlu Yao
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaxing Dai
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bingbing Wu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bo Tian
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiaolin Shi
- Medical College of Soochow University, Suzhou 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
48
|
Luo Z, Qi Z, Luo J, Chen T. Potential applications of engineered bacteria in disease diagnosis and treatment. MICROBIOME RESEARCH REPORTS 2024; 4:10. [PMID: 40207274 PMCID: PMC11977365 DOI: 10.20517/mrr.2024.57] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 04/11/2025]
Abstract
Probiotics are live microorganisms that confer health benefits to the host when administered in appropriate quantities. This beneficial effect has spurred extensive research in the medical and health fields. With rapid advancements in synthetic biology, the genetic and biological characteristics of a broad array of probiotics have been elucidated. Utilizing these insights, genetic editing technologies now enable the precise modification of probiotics, leading to the development of engineered bacteria. Emerging evidence underscores the significant potential of these engineered bacteria in disease management. This review explores the methodologies for creating engineered bacteria, their preliminary applications in healthcare, and the mechanisms underlying their functions. Engineered bacteria are being developed for roles such as in vivo drug delivery systems, biosensors, and mucosal vaccines, thereby contributing to the treatment, diagnosis, and prevention of conditions including inflammatory bowel disease (IBD), metabolic disorders, cancer, and neurodegenerative diseases. The review concludes by assessing the advantages and limitations of engineered bacteria in the context of disease management.
Collapse
Affiliation(s)
- Zhaowei Luo
- School of Huankui Academy, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| | - Zhanghua Qi
- School of Huankui Academy, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| | - Jie Luo
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| |
Collapse
|
49
|
Wang Y, Liu Y, Su X, Niu L, Li N, Xu C, Sun Z, Guo H, Shen S, Yu M. Non-pathogenic Trojan horse Nissle1917 triggers mitophagy through PINK1/Parkin pathway to discourage colon cancer. Mater Today Bio 2024; 29:101273. [PMID: 39415764 PMCID: PMC11480251 DOI: 10.1016/j.mtbio.2024.101273] [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: 04/18/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Bacteria-mediated antitumor therapy has gained widespread attention for its innate tumor-targeting capability and excellent immune activation properties. Nevertheless, the clinical approval of bacterial therapies remains elusive primarily due to the formidable challenge of balancing safety with enhancing in vivo efficacy. In this study, leveraging the probiotic Escherichia coli Nissle1917 (EcN) emerges as a promising approach for colon cancer therapy, offering a high level of safety attributed to its lack of virulence factors and its tumor-targeting potential owing to its obligate anaerobic nature. Specifically, we delineate the erythrocyte (RBC) membrane-camouflaged EcN, termed as Trojan horse EcN@RBC, which triggers apoptosis in tumor cells by mitigating mitochondrial membrane potential (MMP) and subsequently activating the PINK1/Parkin pathway associated with mitophagy. Concurrently, the decline in MMP induced by mitophagy disrupts the mitochondrial permeability transition pore (MPTP), leading to the release of Cytochrome C and subsequent apoptosis induction. Moreover, synergistic effects were observed through the combination of the autophagy activator rapamycin, bolstering the antitumor efficacy in vivo. These findings offer novel insights into probiotic-mediated antitumor mechanisms and underscore the therapeutic potential of EcN@RBC for colon cancer patients.
Collapse
Affiliation(s)
- Yang Wang
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Yao Liu
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaomin Su
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Lili Niu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Nannan Li
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Ce Xu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Zanya Sun
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, Shanghai, 201399, China
| |
Collapse
|
50
|
Liu X, Fan Y, Zhang X, Li L, Yang C, Ma X, Bai G, Sun D, Wang Y, Wang J, Li Y, Shi Y, Liu J, Zhang Y, Wang H. Sequential delivery of photosensitizers and checkpoint inhibitors by engineered bacteria for enhanced cancer photodynamic immunotherapy. Biotechnol Bioeng 2024; 121:3881-3892. [PMID: 39138638 DOI: 10.1002/bit.28829] [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: 04/18/2024] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Engineered bacteria-based cancer therapy has increasingly been considered to be a promising therapeutic strategy due to the development of synthetic biology. Wherein, engineering bacteria-mediated photodynamic therapy (PDT)-immunotherapy shows greater advantages and potential in treatment efficiency than monotherapy. However, the unsustainable regeneration of photosensitizers (PSs) and weak immune responses limit the therapeutic efficiency. Herein, we developed an engineered bacteria-based delivery system for sequential delivery of PSs and checkpoint inhibitors in cancer PDT-immunotherapy. The biosynthetic pathway of 5-aminolevulinic acid (5-ALA) was introduced into Escherichia coli, yielding a supernatant concentration of 172.19 mg/L after 10 h of growth. And another strain was endowed with the light-controllable releasement of anti-programmed cell death-ligand 1 nanobodies (anti-PD-L1). This system exhibited a collaborative effect, where PDT initiated tumor cell death and the released tumor cell fragments stimulated immunity, followed by the elimination of residual tumor cells. The tumor inhibition rate reached 74.97%, and the portion of activated T cells and inflammatory cytokines were reinforced. The results demonstrated that the engineered bacteria-based collaborative system could sequentially deliver therapeutic substance and checkpoint inhibitors, and achieve good therapeutic therapy. This paper will provide a new perspective for the cancer PDT-immunotherapy.
Collapse
Affiliation(s)
- Xinyu Liu
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Yali Fan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Xinyu Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Lianyue Li
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Chao Yang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xiaoyan Ma
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Guijie Bai
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Dawei Sun
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Yaxin Wang
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Junyi Wang
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Yong Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Jing Liu
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| | - Yingying Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Hanjie Wang
- School of Life Sciences, Tianjin University, Tianjin, China
- Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, China
| |
Collapse
|