1
|
Xu H, Yue M, Zhou R, Wang P, Wong MYC, Wang J, Huang H, Chen B, Mo Y, Tam RCY, Zhou B, Du Z, Huang H, Liu L, Tan Z, Yuen KY, Song Y, Chen H, Chen Z. A Prime-Boost Vaccination Approach Induces Lung Resident Memory CD8+ T Cells Derived from Central Memory T Cells That Prevent Tumor Lung Metastasis. Cancer Res 2024; 84:3173-3188. [PMID: 39350665 PMCID: PMC11443216 DOI: 10.1158/0008-5472.can-23-3257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/15/2024] [Accepted: 07/16/2024] [Indexed: 10/04/2024]
Abstract
Memory T cells play a key role in immune protection against cancer. Vaccine-induced tissue-resident memory T (TRM) cells in the lung have been shown to protect against lung metastasis. Identifying the source of lung TRM cells can help to improve strategies, preventing tumor metastasis. Here, we found that a prime-boost vaccination approach using intramuscular DNA vaccine priming, followed by intranasal live-attenuated influenza-vectored vaccine (LAIV) boosting induced higher frequencies of lung CD8+ TRM cells compared with other vaccination regimens. Vaccine-induced lung CD8+ TRM cells, but not circulating memory T cells, conferred significant protection against metastatic melanoma and mesothelioma. Central memory T (TCM) cells induced by the DNA vaccination were major precursors of lung TRM cells established after the intranasal LAIV boost. Single-cell RNA sequencing analysis indicated that transcriptional reprogramming of TCM cells for differentiation into TRM cells in the lungs started as early as day 2 post the LAIV boost. Intranasal LAIV altered the mucosal microenvironment to recruit TCM cells via CXCR3-dependent chemotaxis and induced CD8+ TRM-associated transcriptional programs. These results identified TCM cells as the source of vaccine-induced CD8+ TRM cells that protect against lung metastasis. Significance: Prime-boost vaccination shapes the mucosal microenvironment and reprograms central memory T cells to generate lung resident memory T cells that protect against lung metastasis, providing insights for the optimization of vaccine strategies.
Collapse
Affiliation(s)
- Haoran Xu
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Ming Yue
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
- School of Biomedical Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Runhong Zhou
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Pui Wang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Michael Yik-Chun Wong
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Jinlin Wang
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Huarong Huang
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Bohao Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Yufei Mo
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Rachel Chun-Yee Tam
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Biao Zhou
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Zhenglong Du
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Haode Huang
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Li Liu
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Zhiwu Tan
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Kwok-Yung Yuen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
- Center for Virology, Vaccinology and Therapeutics, Hong Kong, People's Republic of China
| | - Youqiang Song
- School of Biomedical Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Honglin Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
- Center for Virology, Vaccinology and Therapeutics, Hong Kong, People's Republic of China
| | - Zhiwei Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, People's Republic of China
- Center for Virology, Vaccinology and Therapeutics, Hong Kong, People's Republic of China
| |
Collapse
|
2
|
Nguyen DN, Ko P, Roper B, Console G, Gao Y, Shaw D, Yu C, Yehl P, Zhang K, Goyon A. Analytical and Functional Characterization of Plasmid DNA Topological Forms and Multimers. Anal Chem 2024; 96:12801-12808. [PMID: 39051724 DOI: 10.1021/acs.analchem.4c02150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Plasmid DNA (pDNA) is an essential tool in genetic engineering that has gained prevalence in cell and gene therapies. Plasmids exist as supercoiled (SC), open circular (OC), and linear forms. Plasmid multimerization can also occur during the manufacturing process. Even though the SC forms are thought to provide optimal knock-in (KI) efficiency, there is no strong consensus on the effect of the topological forms and multimers on the functional activity. In addition, the results obtained for conventional pDNAs (>5 kbp) do not necessarily translate to smaller pDNAs (∼3 kbp). In this study, a workflow was developed for the analytical and functional characterization of pDNA topological forms and multimers. An anion exchange chromatography (AEC) method was first developed to quantify the topological forms and multimers. Four AEC columns were initially compared, one of which was found to provide superior chromatographic performance. The effect of mobile phase pH, various salts, column temperature, and acetonitrile content on the separation performance was systematically studied. The method performance, including precision and accuracy, was evaluated. The final AEC method was compared to capillary gel electrophoresis (CGE) by analyzing several pDNA sequences and lots. A forced degradation study revealed unexpectedly high degradation of the SC forms. Finally, the KI efficiency was compared for the SC and OC forms, and the multimers.
Collapse
Affiliation(s)
- Daniel Ngoc Nguyen
- Synthetic Molecule Analytical Chemistry, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Peggy Ko
- Cell Therapy Engineering and Development, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Brian Roper
- Cell Therapy Analytical Development, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Gary Console
- Protein Analytical Chemistry, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Yuan Gao
- Cell Therapy Engineering and Development, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - David Shaw
- Cell Therapy Engineering and Development, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Christopher Yu
- Cell Therapy Analytical Development, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Peter Yehl
- Synthetic Molecule Analytical Chemistry, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Kelly Zhang
- Synthetic Molecule Analytical Chemistry, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Alexandre Goyon
- Synthetic Molecule Analytical Chemistry, Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
3
|
Wang XD, Liu YS, Chen MD, Hu MH. Discovery of a triphenylamine-based ligand that targets mitochondrial DNA G-quadruplexes and activates the cGAS-STING immunomodulatory pathway. Eur J Med Chem 2024; 269:116361. [PMID: 38547736 DOI: 10.1016/j.ejmech.2024.116361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/07/2024]
Abstract
Stabilization of G-quadruplex (G4) structures in mitochondria leads to the damage of mitochondrial DNA (mtDNA), making mtDNA G4s a promising target in the field of cancer therapy in recent years. Damaged mtDNA released into the cytosol can stimulate cytosolic DNA-sensing pathways, and cGAS-STING pathway is a typical one with potent immunostimulatory effects. A few small molecule ligands of mtDNA G4s are identified with antitumor efficacy, but little is known about their results and mechanisms on immunomodulation. In this study, we engineered a series of triphenylamine-based analogues targeting mtDNA G4s, and A6 was determined as the most promising compound. Cellular studies indicated that A6 caused severe mtDNA damage. Then, damaged mtDNA stimulated cGAS-STING pathway, resulting in the following cytokine production of tumor cells and the maturation of DCs. In vivo experiments certified that A6 exerted suppressive influences on tumor growth and metastasis in 4T1 cell-bearing mice by the regulation of TME, including the remodeling of macrophages and the activation of T cells. To our knowledge, it is the first time to report a ligand targeting mtDNA G4s to activate the cGAS-STING immunomodulatory pathway, providing a novel strategy for the future development of mtDNA G4-based antitumor agents.
Collapse
Affiliation(s)
- Xiao-Dong Wang
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Yong-Si Liu
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Meng-Die Chen
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Ming-Hao Hu
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China.
| |
Collapse
|
4
|
Cui JZ, Chew ZH, Lim LHK. New insights into nucleic acid sensor AIM2: The potential benefit in targeted therapy for cancer. Pharmacol Res 2024; 200:107079. [PMID: 38272334 DOI: 10.1016/j.phrs.2024.107079] [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: 11/06/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
The AIM2 inflammasome represents a multifaceted oligomeric protein complex within the innate immune system, with the capacity to perceive double-stranded DNA (dsDNA) and engage in diverse physiological reactions and disease contexts, including cancer. While originally conceived as a discerning DNA sensor, AIM2 has demonstrated its capability to discern various nucleic acid variations, encompassing RNA and DNA-RNA hybrids. Through its interaction with nucleic acids, AIM2 orchestrates the assembly of a complex involving multiple proteins, aptly named the AIM2 inflammasome, which facilitates the enzymatic cleavage of proinflammatory cytokines, namely pro-IL-1β and pro-IL-18. This process, in turn, underpins its pivotal biological role. In this review, we provide a systematic summary and discussion of the latest advancements in AIM2 sensing various types of nucleic acids. Additionally, we discuss the modulation of AIM2 activation, which can cause cell death, including pyroptosis, apoptosis, and autophagic cell death. Finally, we fully illustrate the evidence for the dual role of AIM2 in different cancer types, including both anti-tumorigenic and pro-tumorigenic functions. Considering the above information, we uncover the therapeutic promise of modulating the AIM2 inflammasome in cancer treatment.
Collapse
Affiliation(s)
- Jian-Zhou Cui
- Translational Immunology Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore; NUS-Cambridge Immunophenotyping Centre, Life Science Institute, National University of Singapore, Singapore.
| | - Zhi Huan Chew
- Translational Immunology Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lina H K Lim
- Translational Immunology Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
5
|
Raman V, Howell LM, Bloom SMK, Hall CL, Wetherby VE, Minter LM, Kulkarni AA, Forbes NS. Intracellular Salmonella delivery of an exogenous immunization antigen refocuses CD8 T cells against cancer cells, eliminates pancreatic tumors and forms antitumor immunity. Front Immunol 2023; 14:1228532. [PMID: 37868996 PMCID: PMC10585021 DOI: 10.3389/fimmu.2023.1228532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Immunotherapies have shown great promise, but are not effective for all tumors types and are effective in less than 3% of patients with pancreatic ductal adenocarcinomas (PDAC). To make an immune treatment that is effective for more cancer patients and those with PDAC specifically, we genetically engineered Salmonella to deliver exogenous antigens directly into the cytoplasm of tumor cells. We hypothesized that intracellular delivery of an exogenous immunization antigen would activate antigen-specific CD8 T cells and reduce tumors in immunized mice. Methods To test this hypothesis, we administered intracellular delivering (ID) Salmonella that deliver ovalbumin as a model antigen into tumor-bearing, ovalbumin-vaccinated mice. ID Salmonella delivers antigens by autonomously lysing in cells after the induction of cell invasion. Results We showed that the delivered ovalbumin disperses throughout the cytoplasm of cells in culture and in tumors. This delivery into the cytoplasm is essential for antigen cross-presentation. We showed that co-culture of ovalbumin-recipient cancer cells with ovalbumin-specific CD8 T cells triggered a cytotoxic T cell response. After the adoptive transfer of OT-I CD8 T cells, intracellular delivery of ovalbumin reduced tumor growth and eliminated tumors. This effect was dependent on the presence of the ovalbumin-specific T cells. Following vaccination with the exogenous antigen in mice, intracellular delivery of the antigen cleared 43% of established KPC pancreatic tumors, increased survival, and prevented tumor re-implantation. Discussion This response in the immunosuppressive KPC model demonstrates the potential to treat tumors that do not respond to checkpoint inhibitors, and the response to re-challenge indicates that new immunity was established against intrinsic tumor antigens. In the clinic, ID Salmonella could be used to deliver a protein antigen from a childhood immunization to refocus pre-existing T cell immunity against tumors. As an off-the-shelf immunotherapy, this bacterial system has the potential to be effective in a broad range of cancer patients.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Ernest Pharmaceuticals, LLC, Hadley, MA, United States
| | - Lars M. Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
| | - Shoshana M. K. Bloom
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
| | - Christopher L. Hall
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Ernest Pharmaceuticals, LLC, Hadley, MA, United States
| | | | - Lisa M. Minter
- Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
- Institute for Applied Life Science, University of Massachusetts, Amherst, MA, United States
| | - Ashish A. Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Institute for Applied Life Science, University of Massachusetts, Amherst, MA, United States
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA, United States
- Institute for Applied Life Science, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
6
|
Wang XD, Wang JX, Hu MH. Novel phenanthrene imidazoles as telomeric G-quadruplex ligands trigger potent immunogenic cell death in triple-negative breast cancer. Int J Biol Macromol 2023; 249:126068. [PMID: 37524278 DOI: 10.1016/j.ijbiomac.2023.126068] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Immunogenic cell death (ICD) is a typical type of regulated cell demise, and ICD inducers stimulate the immune responses against dead-cell antigens and exert specific antitumor effects. G-quadruplex (G4) binders targeting the telomeres lead to DNA damage response (DDR) and the potential of harnessing the immune system for cancer therapy. However, the immunostimulatory effects of G4 ligands in cancer cells are still seldomly determined. In this study, we rationally designed and synthesized a series of novel phenanthrene imidazoles targeting telomeric G4. Among them, PI-2 was identified as the most promising ligand with high cytotoxicity, cellular uptake efficiency and G4-interacting ability. Cellular studies indicated that PI-2 inhibited the proliferation and migration of both human and mouse triple-negative breast cancer (TNBC) cells. PI-2 triggered the occurrence of DDR and ICD, where the related pathways were further decided. In vivo experiments displayed that PI-2-treated dying cells could be an effective vaccination to reduce tumor burden and promote the infiltration of CD8+ and CD4+ T cells to the tumor microenvironment (TME). To our knowledge, it is the first time to report a DDR-targeted G4 ligand with ICD-inducing ability in immunocompetent animals, which may provide new insights for the development of promising G4-based immunochemotherapeutic agents.
Collapse
Affiliation(s)
- Xiao-Dong Wang
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen 518060, China
| | - Jia-Xin Wang
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen 518060, China
| | - Ming-Hao Hu
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen 518060, China.
| |
Collapse
|
7
|
Luo M, Chen X, Gao H, Yang F, Chen J, Qiao Y. Bacteria-mediated cancer therapy: A versatile bio-sapper with translational potential. Front Oncol 2022; 12:980111. [PMID: 36276157 PMCID: PMC9585267 DOI: 10.3389/fonc.2022.980111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Bacteria are important symbionts for humans, which sustain substantial influences on our health. Interestingly, some bastrains have been identified to have therapeutic applications, notably for antitumor activity. Thereby, oncologists have developed various therapeutic models and investigated the potential antitumor mechanisms for bacteria-mediated cancer therapy (BCT). Even though BCT has a long history and exhibits remarkable therapeutic efficacy in pre-clinical animal models, its clinical translation still lags and requires further breakthroughs. This review aims to focus on the established strains of therapeutic bacteria and their antitumor mechanisms, including the stimulation of host immune responses, direct cytotoxicity, the interference on cellular signal transduction, extracellular matrix remodeling, neoangiogenesis, and metabolism, as well as vehicles for drug delivery and gene therapy. Moreover, a brief discussion is proposed regarding the important future directions for this fantastic research field of BCT at the end of this review.
Collapse
Affiliation(s)
- Miao Luo
- School of Pharmacy, Institute of Hepatology and Metabolic Diseases, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyu Chen
- School of Pharmacy, Institute of Hepatology and Metabolic Diseases, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Haojin Gao
- School of Pharmacy, Institute of Hepatology and Metabolic Diseases, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Fan Yang
- School of Pharmacy, Institute of Hepatology and Metabolic Diseases, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Jianxiang Chen
- School of Pharmacy, Institute of Hepatology and Metabolic Diseases, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiting Qiao, ; Jianxiang Chen,
| | - Yiting Qiao
- School of Pharmacy, Institute of Hepatology and Metabolic Diseases, Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Yiting Qiao, ; Jianxiang Chen,
| |
Collapse
|
8
|
Wu L, Li L, Yin X, Li C, Xin W, Liu L, Hua Z. A SARS-CoV-2 oral vaccine development strategy based on the attenuated Salmonella type III secretion system. J Appl Microbiol 2022; 133:2484-2500. [PMID: 35858677 PMCID: PMC9350170 DOI: 10.1111/jam.15720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/30/2022] [Accepted: 07/14/2022] [Indexed: 11/28/2022]
Abstract
AIMS This study aimed to provide a safe, stable and efficient SARS-CoV-2 oral vaccine development strategy based on the type III secretion system of attenuated Salmonella and a reference for the development of a SARS-CoV-2 vaccine. METHODS AND RESULTS The attenuated Salmonella mutant ΔhtrA-VNP was used as a vector to secrete the antigen SARS-CoV-2 based on the type III secretion system (T3SS). The Salmonella pathogenicity island 2 (SPI-2)-encoded T3SS promoter (sifB) was screened to express heterologous antigens (RBD, NTD, S2), and the SPI-2-encoded secretion system (sseJ) was employed to secrete this molecule (psifB-sseJ-antigen, abbreviated BJ-antigen). Both immunoblotting and fluorescence microscopy revealed effective expression and secretion of the antigen into the cytosol of macrophages in vitro. The mixture of the three strains (BJ-RBD/NTD/S2, named AisVax) elicited a marked increase in the induction of IgA or IgG S-protein Abs after oral gavage, intraperitoneal and subcutaneous administration. Flow cytometric analysis proved that AisVax caused T-cell activation, as shown by a significant increase in CD44 and CD69 expression. Significant production of IgA or IgG N-protein Abs was also detected by using psifB-sseJ-N(FL), indicating the universality of this strategy. CONCLUSIONS Delivery of multiple SARS-CoV-2 antigens using the type III secretion system of attenuated Salmonella ΔhtrA-VNP is a potential COVID-19 vaccine strategy. SIGNIFICANCE AND IMPACT OF THE STUDY The attenuated Salmonella strain ΔhtrA-VNP showed excellent performance as a vaccine vector. The Salmonella SPI-2-encoded T3SS showed highly efficient delivery of SARS-COV-2 antigens. Anti-loss elements integrated into the plasmid stabilized the phenotype of the vaccine strain. Mixed administration of antigen-expressing strains improved antibody induction.
Collapse
Affiliation(s)
- Leyang Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
- Changzhou High‐Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.ChangzhouJiangsuChina
| | - Lin Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
| | - Xingpeng Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
| | - Chenyang Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
| | - Wenjie Xin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
| | - Lina Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingJiangsuChina
- Changzhou High‐Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.ChangzhouJiangsuChina
- School of BiopharmacyChina Pharmaceutical UniversityNanjingJiangsuChina
| |
Collapse
|
9
|
Attenuated Salmonella Typhimurium with truncated LPS and outer membrane-displayed RGD peptide for cancer therapy. Biomed Pharmacother 2022; 155:113682. [PMID: 36095964 DOI: 10.1016/j.biopha.2022.113682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Gram-negative, facultatively anaerobic bacteria Salmonella Typhimurium is a candidate agent or delivery vector for cancer therapy. Effective targeted therapies in addition to radiotherapy, chemotherapy and surgery have been urgently needed as an alternative or supplement. This study expected to further improve the tumor-targeting ability of Salmonella bacteria through genetic modifications. Based on an auxotrophic Salmonella bacterial strain (D2), we constructed Salmonella mutants with altered LPS length to facilitate displaying the RGD4C targeting peptide on the outer membrane surface of Salmonella. The expression of RGD4C peptide in fusion with OmpA was identified by outer membrane protein extraction and WB detection in different mutant strains. However, flow cytometry analysis following immunofluorescence staining demonstrated that the extracellular length of Salmonella LPS did affect the surface display of RGD4C peptide. The strain D2-RGD4C that synthesized intact LPS including lipid A, core oligosaccharides and O antigen polysaccharides could hardly display RGD4C peptide, showing the same fluorescence signal intensity as the strains not expressing RGD4C peptide. Among different strains, D2 ∆rfaJ-RGD4C that synthesized truncated LPS including lipid A and partial core oligosaccharides was capable of displaying RGD4C peptide most efficiently and showed the highest ability to target HUVECs expressing αV integrin and tumor tissue with abundant neovascularization. Animal experiments also demonstrated that this tumor-targeting attenuated Salmonella strain to simultaneously deliver endostatin and TRAIL, two agents with different anti-tumor activities, could significantly inhibit tumor growth and prolong mouse survival. Thus, our studies revealed that Salmonella could be genetically engineered to improve its tumor targeting via the truncation of LPS and surface display of targeting peptides, thereby eliciting superior anti-tumor effects through targeted delivery of drug molecules.
Collapse
|
10
|
Becerra-Báez EI, Meza-Toledo SE, Muñoz-López P, Flores-Martínez LF, Fraga-Pérez K, Magaño-Bocanegra KJ, Juárez-Hernández U, Mateos-Chávez AA, Luria-Pérez R. Recombinant Attenuated Salmonella enterica as a Delivery System of Heterologous Molecules in Cancer Therapy. Cancers (Basel) 2022; 14:cancers14174224. [PMID: 36077761 PMCID: PMC9454573 DOI: 10.3390/cancers14174224] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Cancer is among the main causes of death of millions of individuals worldwide. Although survival has improved with conventional treatments, the appearance of resistant cancer cells leads to patient relapses. It is, therefore, necessary to find new antitumor therapies that can completely eradicate transformed cells. Bacteria-based tumor therapy represents a promising alternative treatment, particularly the use of live-attenuated Salmonella enterica, with its potential use as a delivery system of antitumor heterologous molecules such as tumor-associated antigens, cytotoxic molecules, immunomodulatory molecules, pro-apoptotic proteins, nucleic acids, and nanoparticles. In this review, we present the state of the art of current preclinical and clinical research on the use of Salmonella enterica as a potential therapeutic ally in the war against cancer. Abstract Over a century ago, bacterial extracts were found to be useful in cancer therapy, but this treatment modality was obviated for decades. Currently, in spite of the development and advances in chemotherapies and radiotherapy, failure of these conventional treatments still represents a major issue in the complete eradication of tumor cells and has led to renewed approaches with bacteria-based tumor therapy as an alternative treatment. In this context, live-attenuated bacteria, particularly Salmonella enterica, have demonstrated tumor selectivity, intrinsic oncolytic activity, and the ability to induce innate or specific antitumor immune responses. Moreover, Salmonella enterica also has strong potential as a delivery system of tumor-associated antigens, cytotoxic molecules, immunomodulatory molecules, pro-apoptotic proteins, and nucleic acids into eukaryotic cells, in a process known as bactofection and antitumor nanoparticles. In this review, we present the state of the art of current preclinical and clinical research on the use of Salmonella enterica as a potential therapeutic ally in the war against cancer.
Collapse
Affiliation(s)
- Elayne Irene Becerra-Báez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Sergio Enrique Meza-Toledo
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Paola Muñoz-López
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis Fernando Flores-Martínez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Karla Fraga-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Kevin Jorge Magaño-Bocanegra
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico
| | - Uriel Juárez-Hernández
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Correspondence: ; Tel.: +52-55-52289917 (ext. 4401)
| |
Collapse
|
11
|
Fan JY, Huang Y, Li Y, Muluh TA, Fu SZ, Wu JB. Bacteria in cancer therapy: A new generation of weapons. Cancer Med 2022; 11:4457-4468. [PMID: 35522104 PMCID: PMC9741989 DOI: 10.1002/cam4.4799] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Tumors are presently a major threat to human life and health. Malignant tumors are conventionally treated through radiotherapy and chemotherapy. However, traditional therapies yield unsatisfactory results due to high toxicity to the normal cells, inability to treat deep tumor tissues, and the possibility of inducing drug resistance in the tumor cells. This has caused immunotherapy to emerge as an effective and alternate treatment strategy. To overcome the limitations of the conventional treatments as well as to avert the risk of various drug resistance and cytotoxicity, bacterial anti-tumor immunotherapy has raised the interest of researchers. This therapeutic strategy employs bacteria to specifically target and colonize the tumor tissues with preferential accumulation and proliferation. Such bacterial accumulation initiates a series of anti-tumor immune responses, effectively eliminating the tumor cells. This immunotherapy can use the bacteria alone or concomitantly with the other methods. For example, the bacteria can deliver the anti-cancer effect mediators by regulating the expression of the bacterial genes or by synthesizing the bioengineered bacterial complexes. This review will discuss the mechanism of utilizing bacteria in treating tumors, especially in terms of immune mechanisms. This could help in better integrating the bacterial method with other treatment options, thereby, providing a more effective, reliable, and unique treatment therapy for tumors.
Collapse
Affiliation(s)
- Jun Ying Fan
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Yuan Huang
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Yi Li
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Tobias Achu Muluh
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Shao Zhi Fu
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Department of Nuclear MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Jing Bo Wu
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Department of Nuclear MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Academician (Expert) Workstation of Sichuan ProvinceLuzhouSichuanP.R. China
| |
Collapse
|
12
|
Liu C, Cong X, Wang Y, Guo Q, Xie Y, Geng F, Guo J, Dong L, Zhou Y, Wu H, Yu B, Wu J, Zhang H, Yu X, Kong W. Fast DNA Vaccination Strategy Elicits a Stronger Immune Response Dependent on CD8 +CD11c + Cell Accumulation. Front Oncol 2021; 11:752444. [PMID: 34950581 PMCID: PMC8691261 DOI: 10.3389/fonc.2021.752444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Conventional DNA vaccine strategies usually employ a regimen of immunizations at 2-week or longer intervals to induce effective memory cell-dependent immune responses. Clinical cancer treatment requires a faster immunization strategy to contend with tumor progression. In this study, a novel fast immunization strategy was established, wherein a DNA vaccine was intramuscularly administered on days 0, 2, and 5 in a murine lung cancer model. Effector cells peaked 7 to 10 days after the last vaccination. Compared with traditional 2-week-interval immunization strategies, antigen-specific cytolysis and INF-γ secretion were significantly enhanced under the fast vaccination approach. As a result, the rapidly administered DNA vaccine elicited stronger and more prompt antitumor effects. The probable underlying mechanism of fast immunization was the accumulation of CD8+CD11c+ antigen-presenting cells at the injection site, which enhanced subsequent antigen presentation. In conclusion, the fast DNA vaccination strategy shortened vaccination time to 5 days and elicited a stronger antitumor immune response.
Collapse
Affiliation(s)
- Chenlu Liu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianling Cong
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Yuqian Wang
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Qianqian Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yu Xie
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Fei Geng
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jie Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Ling Dong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yi Zhou
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
13
|
Highlights of Immunomodulation in Salmonella-Based Cancer Therapy. Biomedicines 2021; 9:biomedicines9111566. [PMID: 34829795 PMCID: PMC8615479 DOI: 10.3390/biomedicines9111566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Bacteria-mediated cancer therapy (BMCT) is an emerging tool that may advance potential approaches in cancer immunotherapy, whereby tumors are eradicated by the hosts’ immune system upon recruitment and activation by bacteria such as Salmonella. This paper provides an emphasis on the immunomodulatory effects that encompasses both the innate and adaptive immune responses inherently triggered by Salmonella. Furthermore, modifications of Salmonella-based treatment in the attempt to improve tumor-specific immune responses including cytokine therapy, gene therapy, and DNA vaccine delivery are likewise discussed. The majority of the findings described herein incorporate cell-based experiments and murine model studies, and only a few accounts describe clinical trials. Salmonella-based cancer therapy is still under development; nonetheless, the pre-clinical research and early-phase clinical trials that have been completed so far have shown promising and convincing results. Certainly, the continuous development of, and innovation on, Salmonella-based therapy could pave the way for its eventual emergence as one of the mainstream therapeutic interventions addressing various types of cancer.
Collapse
|
14
|
Hyun J, Jun S, Lim H, Cho H, You SH, Ha SJ, Min JJ, Bang D. Engineered Attenuated Salmonella typhimurium Expressing Neoantigen Has Anticancer Effects. ACS Synth Biol 2021; 10:2478-2487. [PMID: 34525796 DOI: 10.1021/acssynbio.1c00097] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neoantigen vaccines are an immunotherapy strategy for treating cancer. The vaccine degrades quickly, so the strategy must include protection and precise targeting for immune cell stimulation. In this study, we engineered attenuated Salmonella typhimurium, which is highly infiltrative to tumors, to act as a carrier for Neoantigen peptide vaccine. Our system used a constitutive promoter vector, so that a single injection of Salmonella expressing Neoantigen could be used without requiring additional induction injections. In vivo experiments on bacteria-treated mice showed that Neoantigen expressed by the engineered carrier infiltrated tumors and resulted in suppressed tumor growth, higher survival rates and longer survival times, a relative increase of CD4 and CD8 T cells, and cytokine release. These results indicate that engineered Salmonella can be used as a carrier for Neoantigen immunotherapy.
Collapse
Affiliation(s)
- Jungheun Hyun
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Soyeong Jun
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyeonseob Lim
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjun Cho
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung-Hwan You
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam, 58128, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jung-Joon Min
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam, 58128, Republic of Korea
| | - Duhee Bang
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
15
|
Sun H, Li J, Hu W, Yan Y, Guo Z, Zhang Z, Chen Y, Yao X, Teng L, Wang X, Li L, Chai D, Zheng J, Wang G. Co-immunizing with HMGB1 enhances anti-tumor immunity of B7H3 vaccine in renal carcinoma. Mol Immunol 2021; 139:184-192. [PMID: 34560414 DOI: 10.1016/j.molimm.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 11/27/2022]
Abstract
Metastatic renal carcinoma is a kind of tumor with high degree of malignancy, but there are no effective treatment methods and strategies at present. In this study, we designed a folate-grafted PEI600-CyD (H1) nanoparticle-mediated DNA vaccine containing an adjuvant of high mobility group box 1 protein (HMGB1) and a tumor-specific antigen of B7H3 (CD276) for renal carcinoma therapy. Mice bearing subcutaneous human B7H3 (hB7H3)-Renca tumor were immunized with H1-pHMGB1/pB7H3, H1-pB7H3, H1-pHMGB1, or Mock vaccine. Compared to other control groups, the growth of the tumor was significantly inhibited in H1-pHMGB1/pB7H3 vaccine group. The increased proportion and mature of CD11c+ DCs were observed in the spleen of H1-pHMGB1/pB7H3 treated mice. Likewise, HMGB1 promoted B7H3 vaccine to induce tumor-specific CD8+ T cell proliferation and CTL responses. Beyond that, H1-pHMGB1/pB7H3 vaccine strengthened the induction of functional CD8+ T cells. With the depletion of CD8+ T cells, the anti-tumor effect of H1-pHMGB1/pB7H3 also disappeared, indicating that CD8+ T cells are the key factor of the anti-tumor activity of the vaccine. So, to sum up, H1-pHMGB1/pB7H3 vaccine could achieve the desired anti-tumor effect by enhancing the response of tumor-specific functional CD8+ T cell responses. H1 nanoparticle-based vaccines may have great potential and prospect in the treatment of primary solid tumors.
Collapse
Affiliation(s)
- Huanyou Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Juan Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Wenwen Hu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Yinan Yan
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Zengli Guo
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Zichun Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Xuefan Yao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Ling Teng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Xinyuan Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Liantao Li
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| |
Collapse
|
16
|
Howell LM, Forbes NS. Bacteria-based immune therapies for cancer treatment. Semin Cancer Biol 2021; 86:1163-1178. [PMID: 34547442 DOI: 10.1016/j.semcancer.2021.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 12/23/2022]
Abstract
Engineered bacterial therapies that target the tumor immune landscape offer a new class of cancer immunotherapy. Salmonella enterica and Listeria monocytogenes are two species of bacteria that have been engineered to specifically target tumors and serve as delivery vessels for immunotherapies. Therapeutic bacteria have been engineered to deliver cytokines, gene silencing shRNA, and tumor associated antigens that increase immune activation. Bacterial therapies stimulate both the innate and adaptive immune system, change the immune dynamics of the tumor microenvironment, and offer unique strategies for targeting tumors. Bacteria have innate adjuvant properties, which enable both the delivered molecules and the bacteria themselves to stimulate immune responses. Bacterial immunotherapies that deliver cytokines and tumor-associated antigens have demonstrated clinical efficacy. Harnessing the diverse set of mechanisms that Salmonella and Listeria use to alter the tumor-immune landscape has the potential to generate many new and effective immunotherapies.
Collapse
Affiliation(s)
- Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States
| | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States.
| |
Collapse
|
17
|
Cao L, Huang T, Chen X, Li W, Yang X, Zhang W, Li M, Gao R. Uncovering the interplay between pH receptors and immune cells: Potential drug targets (Review). Oncol Rep 2021; 46:228. [PMID: 34476504 DOI: 10.3892/or.2021.8179] [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: 12/30/2020] [Accepted: 05/10/2021] [Indexed: 11/06/2022] Open
Abstract
Extracellular acidosis is associated with various immunopathological states. The microenvironment of numerous solid tumours and inflammatory responses during acute or chronic infection are all related to a pH range of 5.5‑7.0. The relationship between inflammation and immune escape, cancer metabolism, and immunologic suppression drives researchers to focus on the effects of low pH on diverse components of disease immune monitoring. The potential effect of low extracellular pH on the immune function reveals the importance of pH in inflammatory and immunoreactive processes. In this review, the mechanism of how pH receptors, including monocarboxylate transporters (MCTs), Na+/H+ exchanger 1, carbonic anhydrases (CAs), vacuolar‑ATPase, and proton‑sensing G‑protein coupled receptors (GPCRs), modulate the immune system in disease, especially in cancer, were studied. Their role in immunocyte growth and signal transduction as part of the immune response, as well as cytokine production, have been documented in great detail. Currently, immunotherapy strategies have positive therapeutic effects for patients. However, the acidic microenvironment may block the effect of immunotherapy through compensatory feedback mechanisms, leading to drug resistance. Therefore, we highlight promising therapeutic developments regarding pH manipulation and provide a framework for future research.
Collapse
Affiliation(s)
- Lin Cao
- Department of The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100020, P.R. China
| | - Tianqiao Huang
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xiaohong Chen
- Department of Otolaryngology‑Head and Neck Surgery, Beijing Tongren Hospital, Beijing 100010, P.R. China
| | - Weisha Li
- Department of The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100020, P.R. China
| | - Xingjiu Yang
- Department of The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100020, P.R. China
| | - Wenlong Zhang
- Department of The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100020, P.R. China
| | - Mengyuan Li
- Department of The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100020, P.R. China
| | - Ran Gao
- Department of The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100020, P.R. China
| |
Collapse
|
18
|
Yang M, Yang F, Chen W, Liu S, Qiu L, Chen J. Bacteria-mediated cancer therapies: opportunities and challenges. Biomater Sci 2021; 9:5732-5744. [PMID: 34313267 DOI: 10.1039/d1bm00634g] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years, cancer therapy strategies utilizing live tumor-targeting bacteria have presented unique advantages. Engineered bacteria have the particular ability to distinguish tumors from normal tissues with less toxicity. Live bacteria are naturally capable of homing to tumors, resulting in high levels of local colonization because of insufficient oxygen and low pH in the tumor microenvironment. Bacteria initiate their antitumor effects by directly killing the tumor or by activating innate and adaptive antitumor immune responses. The bacterial vectors can be reprogrammed following advanced DNA synthesis, sophisticated genetic bioengineering, and biosensors to engineer microorganisms with complex functions, and then produce and deliver anticancer agents based on clinical needs. However, because of the lack of knowledge on the mechanisms and side effects of microbial cancer therapy, developing such smart microorganisms to treat or prevent cancer remains a significant challenge. In this review, we summarized the potential, status, opportunities and challenges of this growing field. We illustrated the mechanism of tumor regression induced by engineered bacteria and discussed the recent advances in the application of bacteria-mediated cancer therapy to improve efficacy, safety and drug delivery. Finally, we shared our insights into the future directions of tumor-targeting bacteria in cancer therapy.
Collapse
Affiliation(s)
- Meiyang Yang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China.
| | | | | | | | | | | |
Collapse
|
19
|
Chai D, Zhang Z, Jiang N, Ding J, Qiu D, Shi SY, Wang G, Fang L, Li H, Tian H, Yang J, Zhang Q, Zheng J. Co-immunization with L-Myc enhances CD8 + or CD103 + DCs mediated tumor-specific multi-functional CD8 + T cell responses. Cancer Sci 2021; 112:3469-3483. [PMID: 34157192 PMCID: PMC8409417 DOI: 10.1111/cas.15044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/21/2022] Open
Abstract
Renal carcinoma shows a high risk of invasion and metastasis without effective treatment. Herein, we developed a chitosan (CS) nanoparticle-mediated DNA vaccine containing an activated factor L-Myc and a tumor-specific antigen CAIX for renal carcinoma treatment. The subcutaneous tumor models were intramuscularly immunized with CS-pL-Myc/pCAIX or control vaccine, respectively. Compared with single immunization group, the tumor growth was significantly suppressed in CS-pL-Myc/pCAIX co-immunization group. The increased proportion and mature of CD11c+ DCs, CD8+ CD11c+ DCs and CD103+ CD11c+ DCs were observed in the splenocytes from CS-pL-Myc/pCAIX co-immunized mice. Furthermore, the enhanced antigen-specific CD8+ T lymphocyte proliferation, cytotoxic T lymphocyte (CTL) responses, and multi-functional CD8+ T cell induction were detected in CS-pL-Myc/pCAIX co-immunization group compared with CS-pCAIX immunization group. Of note, the depletion of CD8 T cells resulted in the reduction of CD8+ T cells or CD8+ CD11c+ DCs and the loss of anti-tumor efficacy induced by CS-pL-Myc/pCAIX vaccine, suggesting the therapeutic efficacy of the vaccine was required for CD8+ DCs and CD103+ DCs mediated CD8+ T cells responses. Likewise, CS-pL-Myc/pCAIX co-immunization also significantly inhibited the lung metastasis of renal carcinoma models accompanied with the increased induction of multi-functional CD8+ T cell responses. Therefore, these results indicated that CS-pL-Myc/pCAIX vaccine could effectively induce CD8+ DCs and CD103+ DCs mediated tumor-specific multi-functional CD8+ T cell responses and exert the anti-tumor efficacy. This vaccine strategy offers a potential and promising approach for solid or metastatic tumor treatment.
Collapse
Affiliation(s)
- Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zichun Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Nan Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiage Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dong Qiu
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shang Yuchen Shi
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Tian
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jie Yang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
20
|
Lin Q, Rong L, Jia X, Li R, Yu B, Hu J, Luo X, Badea SR, Xu C, Fu G, Lai K, Lee MC, Zhang B, Gong H, Zhou N, Chen XL, Lin SH, Fu G, Huang JD. IFN-γ-dependent NK cell activation is essential to metastasis suppression by engineered Salmonella. Nat Commun 2021; 12:2537. [PMID: 33953170 PMCID: PMC8099885 DOI: 10.1038/s41467-021-22755-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Metastasis accounts for 90% of cancer-related deaths and, currently, there are no effective clinical therapies to block the metastatic cascade. A need to develop novel therapies specifically targeting fundamental metastasis processes remains urgent. Here, we demonstrate that Salmonella YB1, an engineered oxygen-sensitive strain, potently inhibits metastasis of a broad range of cancers. This process requires both IFN-γ and NK cells, as the absence of IFN-γ greatly reduces, whilst depletion of NK cells in vivo completely abolishes, the anti-metastatic ability of Salmonella. Mechanistically, we find that IFN-γ is mainly produced by NK cells during early Salmonella infection, and in turn, IFN-γ promotes the accumulation, activation, and cytotoxicity of NK cells, which kill the metastatic cancer cells thus achieving an anti-metastatic effect. Our findings highlight the significance of a self-regulatory feedback loop of NK cells in inhibiting metastasis, pointing a possible approach to develop anti-metastatic therapies by harnessing the power of NK cells.
Collapse
Affiliation(s)
- Qiubin Lin
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,HKU-Zhejiang Institute of Research and Innovation (HKU-ZIRI), Hangzhou, China
| | - Li Rong
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Xian Jia
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Renhao Li
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Bin Yu
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Jingchu Hu
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiao Luo
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - S. R. Badea
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Chen Xu
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Guofeng Fu
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Kejiong Lai
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Ming-chun Lee
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Baozhong Zhang
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huarui Gong
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Nan Zhou
- grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiao Lei Chen
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China ,grid.12955.3a0000 0001 2264 7233Cancer Research Center of Xiamen University, Xiamen, China
| | - Shu-hai Lin
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China
| | - Guo Fu
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Medicine, Xiamen University, Xiamen, China ,grid.12955.3a0000 0001 2264 7233Cancer Research Center of Xiamen University, Xiamen, China
| | - Jian-Dong Huang
- grid.194645.b0000000121742757School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,HKU-Zhejiang Institute of Research and Innovation (HKU-ZIRI), Hangzhou, China ,grid.9227.e0000000119573309Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
21
|
Liu Y, Tang L, Gao N, Diao Y, Zhong J, Deng Y, Wang Z, Jin G, Wang X. Synthetic MUC1 breast cancer vaccine containing a Toll-like receptor 7 agonist exerts antitumor effects. Oncol Lett 2020; 20:2369-2377. [PMID: 32782554 PMCID: PMC7400475 DOI: 10.3892/ol.2020.11762] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
Adjuvant immunotherapy has recently emerged as a potential treatment strategy for breast cancer. The tumor-associated protein mucin 1 (MUC1) has received increasing attention due to its high expression in numerous types of common tumors, in which MUC1 acts as a cancer antigen. However, the simple mixed composition of an adjuvant and a peptide is not a sufficient rationale for a MUC1 peptide-based vaccine. The present study developed a novel Toll-like receptor 7 (TLR7) agonist-conjugated MUC1 peptide vaccine (T7-MUC1), which elicited an effective immune response and a robust antitumor effect in a mouse breast cancer model. In vitro, T7-MUC1 significantly increased the release of cytokines in mouse bone marrow dendritic cells and spleen lymphocytes, and induced the dendritic cell-cytokine-induced killer response against tumor cells with high MUC1 expression. In vivo, it was observed that the 4T1 tumor weights in mice immunized with the T7-MUC1 conjugate were reduced by ≥70% compared with those in the control group. Furthermore, the therapeutic responses in vivo were attributed to the increase in specific humoral and cellular immunity, including high antibody titers, antibody-dependent cell-mediated cytotoxicity and cytotoxic T-lymphocyte activity. The percentages of CD3+/CD8+ T-cells were significantly higher in the T7-MUC1 treatment group compared with those in the control group. Therefore, the results of the present study suggested that the T7-MUC1 vaccine inhibited tumor growth in mice and thus may have potential as a therapeutic candidate in clinical trials for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Yu Liu
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China.,Department of Research and Education, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518001, P.R. China
| | - Li Tang
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China.,College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of The Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Ningning Gao
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Yuwen Diao
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Jingjing Zhong
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Yongqiang Deng
- Department of Oral and Maxillofacial Surgery, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Zhulin Wang
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Guangyi Jin
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Xiaodong Wang
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| |
Collapse
|
22
|
Guo S, Xiao P, Li B, Wang W, Wang S, Lv T, Xu X, Chen C, Huang L, Li Z, Tang L, Peng L, Wang H. Co-immunizing with PD-L1 induces CD8 + DCs-mediated anti-tumor immunity in multiple myeloma. Int Immunopharmacol 2020; 84:106516. [PMID: 32334387 DOI: 10.1016/j.intimp.2020.106516] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/24/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022]
Abstract
Tumor therapeutic vaccines have faced a challenge for effective protection against malignant tumors by inducing tumor-specific CD8+ T cell responses. Here, we designed a DNA vaccine containing a tumor-specific antigen of Dickkopf-1 (DKK-1) and an immune checkpoint of programmed death ligand 1 (PD-L1) delivered by PLGA/PEI nanoparticle-mediated delivery system for multiple myeloma therapy. Murine subcutaneous tumor model established with human DKK1 (hDKK-1)-SP2/0 cells were intramuscularly immunized with PLGA/PEI-pPD-L1/pDDK-1 vaccine and equal amount of control 3 times at 10 day-intervals. Compared with PLGA/PEI-pDKK1 immunization group, PLGA/PEI-pPD-L1/pDKK-1 co-immunization enhanced the induction and mature of CD11c+ DCs and CD8+CD11c+ DCs, and promoted antigen-specific Th1 responses and cytotoxic T lymphocyte (CTL) responses. The reduced tumor volume and weight as well as increased tumor inhibition rate were observed in PLGA/PEI-pPD-L1/pDKK-1 vaccine co-immunization group, indicated that the vaccine could effectively inhibit the tumor growth of multiple myeloma. The anti-tumor activity of PLGA/PEI-pPD-L1/pDKK-1 vaccine was abrogated by CD8 cell depletion accompanied with the reduced percentages of CD8+CD11c+ DCs and CD8+ T cells in the spleen and TILs. These results indicated that the anti-tumor efficacy of PLGA/PEI-pPD-L1/pDKK-1 vaccine was required for CD8+CD11c+ DCs-mediated CD8+ T cell immunity responses. This vaccine strategy may represent a potential and promising approach for hematological malignancy treatment.
Collapse
Affiliation(s)
- Shuli Guo
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Pengli Xiao
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Bo Li
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Wanli Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Songyun Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Tao Lv
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Xiaoyan Xu
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Cong Chen
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Lei Huang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Zhi Li
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Li Tang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Liang Peng
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Huirui Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China.
| |
Collapse
|
23
|
Wang M, Qazi IH, Wang L, Zhou G, Han H. Salmonella Virulence and Immune Escape. Microorganisms 2020; 8:microorganisms8030407. [PMID: 32183199 PMCID: PMC7143636 DOI: 10.3390/microorganisms8030407] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Salmonella genus represents the most common foodborne pathogens causing morbidity, mortality, and burden of disease in all regions of the world. The introduction of antimicrobial agents and Salmonella-specific phages has been considered as an effective intervention strategy to reduce Salmonella contamination. However, data from the United States, European countries, and low- and middle-income countries indicate that Salmonella cases are still a commonly encountered cause of bacterial foodborne diseases globally. The control programs have not been successful and even led to the emergence of some multidrug-resistant Salmonella strains. It is known that the host immune system is able to effectively prevent microbial invasion and eliminate microorganisms. However, Salmonella has evolved mechanisms of resisting host physical barriers and inhibiting subsequent activation of immune response through their virulence factors. There has been a high interest in understanding how Salmonella interacts with the host. Therefore, in the present review, we characterize the functions of Salmonella virulence genes and particularly focus on the mechanisms of immune escape in light of evidence from the emerging mainstream literature.
Collapse
Affiliation(s)
- Mengyao Wang
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Izhar Hyder Qazi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
- Department of Veterinary Anatomy and Histology, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan
| | - Linli Wang
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guangbin Zhou
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
- Correspondence: (H.H.); (G.Z.)
| | - Hongbing Han
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- Correspondence: (H.H.); (G.Z.)
| |
Collapse
|
24
|
Guo Y, Chen Y, Liu X, Min JJ, Tan W, Zheng JH. Targeted cancer immunotherapy with genetically engineered oncolytic Salmonella typhimurium. Cancer Lett 2020; 469:102-110. [DOI: 10.1016/j.canlet.2019.10.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/29/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022]
|
25
|
Broadway KM, Scharf BE. Salmonella Typhimurium as an Anticancer Therapy: Recent Advances and Perspectives. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019. [DOI: 10.1007/s40588-019-00132-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
26
|
Mi Z, Feng ZC, Li C, Yang X, Ma MT, Rong PF. Salmonella-Mediated Cancer Therapy: An Innovative Therapeutic Strategy. J Cancer 2019; 10:4765-4776. [PMID: 31598148 PMCID: PMC6775532 DOI: 10.7150/jca.32650] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/06/2019] [Indexed: 12/31/2022] Open
Abstract
Bacterial-mediated cancer therapy (BMCT) has become a hot topic in the area of antitumor treatment. Salmonella has been recommended to specifically colonize and proliferate inside tumors and even inhibit tumor growth. Salmonella typhimurium (S. typhimurium) is one of the most promising mediators, which can be easily manipulated. S. typhimurium has been engineered and designed as cancer-targeting therapeutics, and can be improved by combining with other therapeutic methods, e.g. chemotherapy and radiotherapy, which regulate the tumor microenvironment synergistically. In view of all these strengths, the engineered attenuated strains have significant advantages for tumor diagnosis and treatment. This treatment has also been approved by the FDA for clinical trial. In this review, we summarized the recent progress and research in the field of Salmonella -mediated cancer therapy.
Collapse
Affiliation(s)
- Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zhi-Chao Feng
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Cheng Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Xiao Yang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Meng-Tian Ma
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Peng-Fei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
27
|
Xu X, Yi C, Yang X, Xu J, Sun Q, Liu Y, Zhao L. Tumor Cells Modified with Newcastle Disease Virus Expressing IL-24 as a Cancer Vaccine. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:213-221. [PMID: 31338417 PMCID: PMC6630061 DOI: 10.1016/j.omto.2019.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/04/2019] [Indexed: 12/24/2022]
Abstract
Interleukin-24 (IL-24) is a promising agent for cancer immunotherapy that induces apoptosis of tumor cells and enhances T cell activation and function. In order to improve the antitumor activity induced by Newcastle disease virus (NDV)-modified tumor vaccine, we generated a recombinant NDV expressing IL-24 using reverse genetics. Irradiated tumor cells infected with LX/IL-24 showed stable IL-24 expression. The cytotoxicity assay showed that LX/IL-24-infected murine melanoma cells significantly enhanced the antitumor immune response in vitro. Then, the antitumor effects of virus-infected tumor cells were examined in the murine tumor models. LX/IL-24-infected tumor cells exhibited strong antitumor effects both in prophylaxis and therapeutic models. LX/IL-24-infected tumor cells increased infiltration of CD4+ T cells and CD8+ T cells in tumor sites, and the antitumor activity of the tumor vaccine modified with LX/IL-24 was dependent on CD8+ T cells. Taken together, our data well illustrates that LX/IL-24-modified tumor cells are a promising agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaojing Xu
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, China
| | - Cheng Yi
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, China
| | - Xiaoqin Yang
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, China
| | - Jianwei Xu
- National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, 550004 Guiyang, Guizhou, China.,Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou, China
| | - Qing Sun
- Laboratory of Animal Infectious Diseases, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.,Virus Research Unit, Department of Microbiology and Immunology, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Yonghao Liu
- Institute of Blood and Marrow Transplantation, Department of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, 215123 Suzhou, People's Republic of China
| | - Lixiang Zhao
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123 Suzhou, China
| |
Collapse
|
28
|
Luo L, Lv M, Zhuang X, Zhang Q, Qiao T. Irradiation increases the immunogenicity of lung cancer cells and irradiation-based tumor cell vaccine elicits tumor-specific T cell responses in vivo. Onco Targets Ther 2019; 12:3805-3815. [PMID: 31190880 PMCID: PMC6529730 DOI: 10.2147/ott.s197516] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 03/08/2019] [Indexed: 12/21/2022] Open
Abstract
Background: During the past decades, great efforts have been built to develop lung cancer vaccines. Whole tumor cell lysate (TCL) are ideal sources of antigens for cancer vaccine design, which however have limited efficacy due to insufficient immunogenicity. Recently, radiotherapy has been closely related to immunotherapy. Numerous studies have demonstrated the regulatory effect of irradiation (IR) on tumor immune response. Purpose: To explore the immunogenicity modulation effect of IR on lung cancer cells. Methods: RNA-sequence and qPCR assay was used to evaluate the change of tumor antigens expression after repeated X rays radiation on A549 cells. Vaccine based on TCL of irradiated Lewis lung cancer cells (IR-LLC) was established; therapeutic effect of TCL (IR-LLC) was examined in xenografted tumor model of mice. Flow cytometry was conducted to evaluate the rate of immune cells in spleen; ELISA was used to detect the level of cytokines in plasma. Immunohistochemistry was performed to evaluate the infiltrations of T-cell in tumor tissues; TIMER analysis was used to explore the correlations between tumor antigen expressions and the abundances of immune infiltrates. Results: IR upregulated the expression of tumor antigens in A549 cells. Compared to the control group and unirradiated tumor cell vaccine, TCL(IR-LLC) had a significantly stronger anti-tumor effect in mice bearing with LLC xenografts. TCL(IR-LLC) significantly increased matured DCs and total CD4+ T cells but downregulated Tregs and PD-1+ CD8+ T cells in mice spleen; TCL(IR-LLC) vaccine upregulated the level of IFN-γ and IL-4 while decreased IL-10 in serum; increased infiltrations of CD4+ T-cells and CD8+ T-cells were observed in the tumor issues of mice immunized with TCL(IR-LLC). Tumor antigens including FN1, MFGE8, MMP2, MYL9 may contribute to the enhanced T-cell response. Conclusion: This study confirmed the immunogenicity modulation effect of IR in NSCLC cells, indicating IR might be an effective strategy to enhance the anti-tumor immunity of cancer cell vaccine.
Collapse
Affiliation(s)
- Lumeng Luo
- Department of Oncology, Jinshan Hospital, Medical Center of Fudan University, Shanghai201500, People’s Republic of China
| | - Minghe Lv
- Department of Oncology, Jinshan Hospital, Medical Center of Fudan University, Shanghai201500, People’s Republic of China
| | - Xibing Zhuang
- Department of Oncology, Jinshan Hospital, Medical Center of Fudan University, Shanghai201500, People’s Republic of China
| | - Qi Zhang
- Department of Oncology, Jinshan Hospital, Medical Center of Fudan University, Shanghai201500, People’s Republic of China
| | - Tiankui Qiao
- Department of Oncology, Jinshan Hospital, Medical Center of Fudan University, Shanghai201500, People’s Republic of China
| |
Collapse
|
29
|
Liang K, Liu Q, Li P, Luo H, Wang H, Kong Q. Genetically engineered Salmonella Typhimurium: Recent advances in cancer therapy. Cancer Lett 2019; 448:168-181. [PMID: 30753837 DOI: 10.1016/j.canlet.2019.01.037] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 12/13/2022]
Abstract
Bacteria have been investigated as anti-tumor therapeutic agents for more than a century, since Coley first observed successful curing of a patient with inoperable cancer by injection of streptococcal organisms. Previous studies have demonstrated that some obligate or facultative anaerobes can selectively accumulate and proliferate within tumors and suppress their growth. Developments in molecular biology as well as the complete genome sequencing of many bacterial species have increased the applicability of bacterial organisms for cancer treatment. In particular, the facultative anaerobe Salmonella Typhimurium has been widely studied and genetically engineered to improve its tumor-targeting ability as well as to reduce bacterial virulence. Moreover, the effectiveness of engineered attenuated S. Typhimurium strains employed as live delivery vectors of various anti-tumor therapeutic agents or combined with other therapies has been evaluated in a large number of animal experiments. The well-known S. Typhimurium mutant VNP20009 and its derivative strain TAPET-CD have even been applied in human clinical trials. However, Salmonella-mediated cancer therapies have not achieved the expected success, except in animal experiments. Many problems remain to be solved to exploit more promising strategies for combatting cancer with Salmonella bacteria. Here, we summarize the promising studies regarding cancer therapy mediated by Salmonella bacteria and highlight the main mechanisms of Salmonella anti-tumor activities.
Collapse
Affiliation(s)
- Kang Liang
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Qing Liu
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Pei Li
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Hongyan Luo
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Haoju Wang
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Qingke Kong
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China; Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, 32608, USA.
| |
Collapse
|
30
|
Carbohydrate-based adjuvants activate tumor-specific Th1 and CD8+ T-cell responses and reduce the immunosuppressive activity of MDSCs. Cancer Lett 2019; 440-441:94-105. [DOI: 10.1016/j.canlet.2018.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/24/2018] [Accepted: 10/10/2018] [Indexed: 01/09/2023]
|
31
|
Arora D, Sharma C, Jaglan S, Lichtfouse E. Live-Attenuated Bacterial Vectors for Delivery of Mucosal Vaccines, DNA Vaccines, and Cancer Immunotherapy. ENVIRONMENTAL CHEMISTRY FOR A SUSTAINABLE WORLD 2019. [PMCID: PMC7123696 DOI: 10.1007/978-3-030-01881-8_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vaccines save millions of lives each year from various life-threatening infectious diseases, and there are more than 20 vaccines currently licensed for human use worldwide. Moreover, in recent decades immunotherapy has become the mainstream therapy, which highlights the tremendous potential of immune response mediators, including vaccines for prevention and treatment of various forms of cancer. However, despite the tremendous advances in microbiology and immunology, there are several vaccine preventable diseases which still lack effective vaccines. Classically, weakened forms (attenuated) of pathogenic microbes were used as vaccines. Although the attenuated microbes induce effective immune response, a significant risk of reversion to pathogenic forms remains. While in the twenty-first century, with the advent of genetic engineering, microbes can be tailored with desired properties. In this review, I have focused on the use of genetically modified bacteria for the delivery of vaccine antigens. More specifically, the live-attenuated bacteria, derived from pathogenic bacteria, possess many features that make them highly suitable vectors for the delivery of vaccine antigens. Bacteria can theoretically express any heterologous gene or can deliver mammalian expression vectors harboring vaccine antigens (DNA vaccines). These properties of live-attenuated microbes are being harnessed to make vaccines against several infectious and noninfectious diseases. In this regard, I have described the desired features of live-attenuated bacterial vectors and the mechanisms of immune responses manifested by live-attenuated bacterial vectors. Interestingly anaerobic bacteria are naturally attracted to tumors, which make them suitable vehicles to deliver tumor-associated antigens thus I have discussed important studies investigating the role of bacterial vectors in immunotherapy. Finally, I have provided important discussion on novel approaches for improvement and tailoring of live-attenuated bacterial vectors for the generation of desired immune responses.
Collapse
Affiliation(s)
- Divya Arora
- Indian Institute of Integrative Medicine, CSIR, Jammu, India
| | - Chetan Sharma
- Guru Angad Dev Veterinary and Animal Science University, Ludhiana, Punjab India
| | - Sundeep Jaglan
- Indian Institute of Integrative Medicine, CSIR, Jammu, India
| | - Eric Lichtfouse
- Aix Marseille University, CNRS, IRD, INRA, Coll France, CEREGE, Aix en Provence, France
| |
Collapse
|
32
|
Jin Z, Lei L, Lin D, Liu Y, Song Y, Gong H, Zhu Y, Mei Y, Hu B, Wu Y, Zhang G, Liu H. IL-33 Released in the Liver Inhibits Tumor Growth via Promotion of CD4 + and CD8 + T Cell Responses in Hepatocellular Carcinoma. THE JOURNAL OF IMMUNOLOGY 2018; 201:3770-3779. [PMID: 30446569 DOI: 10.4049/jimmunol.1800627] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 10/17/2018] [Indexed: 12/28/2022]
Abstract
IL-33 released by epithelial cells and immune cells functions as an alarmin and can induce both type 1 and type 2 immune responses. However, the role of IL-33 release in tumor development is still not clear. In this study, we examined the function of released IL-33 in murine hepatocellular carcinoma (HCC) models by hydrodynamically injecting either IL-33-expressing tumor cells or IL-33-expressing plasmids into the liver of tumor-bearing mice. Tumor growth was greatly inhibited by IL-33 release. This antitumor effect of IL-33 was dependent on suppression of tumorigenicity 2 (ST2) because it was diminished in ST2-/- mice. Moreover, HCC patients with high IL-33 expression have prolonged overall survival compared with the patients with low IL-33 expression. Further study showed that there were increased percentages and numbers of activated and effector CD4+ and CD8+ T cells in both spleen and liver in IL-33-expressing tumor-bearing mice. Moreover, IFN-γ production of the CD4+ and CD8+ T cells was upregulated in both spleen and liver by IL-33. The cytotoxicity of CTLs from IL-33-expressing mice was also enhanced. In vitro rIL-33 treatment could preferentially expand CD8+ T cells and promote CD4+ and CD8+ T cell activation and IFN-γ production. Depletion of CD4+ and CD8+ T cells diminished the antitumor activity of IL-33, suggesting that the antitumor function of released IL-33 was mediated by both CD4+ and CD8+ T cells. Taken together, we demonstrated in murine HCC models that IL-33 release could inhibit tumor development through its interaction with ST2 to promote antitumor CD4+ and CD8+ T cell responses.
Collapse
Affiliation(s)
- Ziqi Jin
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Lei Lei
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Dandan Lin
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Yonghao Liu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Yuan Song
- Immunology Program, Department of Microbiology and Immunology, National University of Singapore, Singapore 117456, Singapore.,Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Ying Zhu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Yu Mei
- Immunology Program, Department of Microbiology and Immunology, National University of Singapore, Singapore 117456, Singapore.,Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Yan Wu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University-, Medical College, Soochow University, Suzhou 215123, China
| | - Guangbo Zhang
- Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; and.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Haiyan Liu
- Immunology Program, Department of Microbiology and Immunology, National University of Singapore, Singapore 117456, Singapore; .,Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
33
|
Chai D, Shan H, Wang G, Zhang Q, Li H, Fang L, Song J, Liu N, Zhang Q, Yao H, Zheng J. Combining DNA Vaccine and AIM2 in H1 Nanoparticles Exert Anti-Renal Carcinoma Effects via Enhancing Tumor-Specific Multi-functional CD8 + T-cell Responses. Mol Cancer Ther 2018; 18:323-334. [PMID: 30401695 DOI: 10.1158/1535-7163.mct-18-0832] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/02/2018] [Accepted: 10/31/2018] [Indexed: 11/16/2022]
Abstract
Renal carcinoma presents a rapid progression in patients with high metastasis with no effective therapeutic strategy. In this study, we designed a folate-grafted PEI600-CyD (H1) nanoparticle-mediated DNA vaccine containing an adjuvant of absent in melanoma 2 (AIM2) and a tumor-specific antigen of carbonic anhydrase IX (CAIX) for renal carcinoma therapy. Mice bearing subcutaneous human CAIX (hCAIX)-Renca tumor were intramuscularly immunized with H1-pAIM2/pCAIX, H1-pCAIX, H1-pAIM2, or Mock vaccine, respectively. The tumor growth of hCAIX-Renca was significantly inhibited in H1-pAIM2/pCAIX vaccine group compared with the control group. The vaccine activated CAIX-specific CD8+ T-cell proliferation and CTL responses, and enhanced the induction of multi-functional CD8+ T cells (expressing TNF-α, IL-2, and IFN-γ). CD8+ T-cell depletion resulted in the loss of anti-tumor activity of H1-pAIM2/pCAIX vaccine, suggesting that the efficacy of the vaccine was dependent on CD8+ T-cell responses. Lung metastasis of renal carcinoma was also suppressed by H1-pAIM2/pCAIX vaccine treatment accompanied with the increased percentages of CAIX-specific multi-functional CD8+ T cells in the spleen, tumor, and bronchoalveolar lavage as compared with H1-pCAIX vaccine. Similarly, the vaccine enhanced CAIX-specific CD8+ T-cell proliferation and CTL responses. Therefore, these results indicated that H1-pAIM2/pCAIX vaccine exhibits the therapeutic efficacy of anti-renal carcinoma by enhancing tumor-specific multi-functional CD8+ T-cell responses. This vaccine strategy could be a potential and promising approach for the therapy of primary solid or metastasis tumors.
Collapse
Affiliation(s)
- Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongjian Shan
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jingyuan Song
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Nianli Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qian Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Yao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
34
|
Farris E, Sanderfer K, Lampe A, Brown DM, Ramer-Tait AE, Pannier AK. Oral Non-Viral Gene Delivery for Applications in DNA Vaccination and Gene Therapy. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 7:51-57. [PMID: 31011691 PMCID: PMC6474414 DOI: 10.1016/j.cobme.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Non-viral gene delivery via the oral route is a promising strategy for improving outcomes of DNA vaccination and gene therapy applications. Unlike traditional parenteral administration routes, the oral route is a non-invasive approach that lends itself to high patient compliance and ease of dosing. Moreover, oral administration allows for both local and systemic production of therapeutic genes or, in the case of DNA vaccination, mucosal and systemic immunity. However, the oral route presents distinct challenges and barriers to achieving successful gene delivery. Oral non-viral gene delivery systems must be able to survive the harsh and variable environments (e.g. acidic pH, degrading enzymes, mucus layer) encountered during transit through the gastrointestinal tract, while still allowing for efficient transgene production at sites of interest. These barriers present unique design challenges for researchers in material selection and in improving the transfection efficiency of orally delivered genes. This review provides an overview of advancements in the design of oral non-viral gene delivery systems, and highlights recent and important developments towards improving orally delivered genes for applications in gene therapy and DNA vaccination.
Collapse
Affiliation(s)
- Eric Farris
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Kari Sanderfer
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Anna Lampe
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583
- Nebraska Center for Materials and Nanoscience, University of Nebraska-Lincoln, Lincoln, NE 68588
- Center for Nanohybrid Functional Materials, University of Nebraska-Lincoln, Lincoln, NE 68588
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
35
|
Pierini S, Perales-Linares R, Uribe-Herranz M, Pol JG, Zitvogel L, Kroemer G, Facciabene A, Galluzzi L. Trial watch: DNA-based vaccines for oncological indications. Oncoimmunology 2017; 6:e1398878. [PMID: 29209575 DOI: 10.1080/2162402x.2017.1398878] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/16/2022] Open
Abstract
DNA-based vaccination is a promising approach to cancer immunotherapy. DNA-based vaccines specific for tumor-associated antigens (TAAs) are indeed relatively simple to produce, cost-efficient and well tolerated. However, the clinical efficacy of DNA-based vaccines for cancer therapy is considerably limited by central and peripheral tolerance. During the past decade, considerable efforts have been devoted to the development and characterization of novel DNA-based vaccines that would circumvent this obstacle. In this setting, particular attention has been dedicated to the route of administration, expression of modified TAAs, co-expression of immunostimulatory molecules, and co-delivery of immune checkpoint blockers. Here, we review preclinical and clinical progress on DNA-based vaccines for cancer therapy.
Collapse
Affiliation(s)
- Stefano Pierini
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renzo Perales-Linares
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mireia Uribe-Herranz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan G Pol
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Andrea Facciabene
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|