1
|
Kuru-Yaşar R, Üstün-Aytekin Ö. The Crucial Roles of Diet, Microbiota, and Postbiotics in Colorectal Cancer. Curr Nutr Rep 2024; 13:126-151. [PMID: 38483752 PMCID: PMC11133122 DOI: 10.1007/s13668-024-00525-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF REVIEW Colorectal cancer is the second deadliest cancer in the world, and its prevalence has been increasing alarmingly in recent years. After researchers discovered the existence of dysbiosis in colorectal cancer, they considered the use of probiotics in the treatment of colorectal cancer. However, for various reasons, including the low safety profile of probiotics in susceptible and immunocompromised patient5s, and the risk of developing antibiotic resistance, researchers have shifted their focus to non-living cells, their components, and metabolites. This review aims to comprehensively evaluate the literature on the effects of diet, microbiota, and postbiotics on colorectal cancer and the future of postbiotics. RECENT FINDINGS The link between diet, gut microbiota, and colorectal cancer has been established primarily as a relationship rather than a cause-effect relationship. The gut microbiota can convert gastrointestinal tract and dietary factors into either onco-metabolites or tumor suppressor metabolites. There is serious dysbiosis in the microbiota in colorectal cancer. Postbiotics appear to be promising agents in the prevention and treatment of colorectal cancer. It has been shown that various postbiotics can selectively induce apoptosis in CRC, inhibit cell proliferation, growth, invasion, and migration, modulate the immune system, suppress carcinogenic signaling pathways, maintain intestinal epithelial integrity, and have a synergistic effect with chemotherapy drugs. However, it is also reported that some postbiotics are ineffective and may be risky in terms of safety profile in some patients. Many issues need to be researched about postbiotics. Large-scale, randomized, double-blind clinical studies are needed.
Collapse
Affiliation(s)
- Rüya Kuru-Yaşar
- Department of Nutrition and Dietetics, Hamidiye Faculty of Health Sciences, University of Health Sciences, 34668, Istanbul, Türkiye
| | - Özlem Üstün-Aytekin
- Department of Nutrition and Dietetics, Hamidiye Faculty of Health Sciences, University of Health Sciences, 34668, Istanbul, Türkiye.
| |
Collapse
|
2
|
Roe JM, Seely K, Bussard CJ, Eischen Martin E, Mouw EG, Bayles KW, Hollingsworth MA, Brooks AE, Dailey KM. Hacking the Immune Response to Solid Tumors: Harnessing the Anti-Cancer Capacities of Oncolytic Bacteria. Pharmaceutics 2023; 15:2004. [PMID: 37514190 PMCID: PMC10384176 DOI: 10.3390/pharmaceutics15072004] [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: 06/26/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Oncolytic bacteria are a classification of bacteria with a natural ability to specifically target solid tumors and, in the process, stimulate a potent immune response. Currently, these include species of Klebsiella, Listeria, Mycobacteria, Streptococcus/Serratia (Coley's Toxin), Proteus, Salmonella, and Clostridium. Advancements in techniques and methodology, including genetic engineering, create opportunities to "hijack" typical host-pathogen interactions and subsequently harness oncolytic capacities. Engineering, sometimes termed "domestication", of oncolytic bacterial species is especially beneficial when solid tumors are inaccessible or metastasize early in development. This review examines reported oncolytic bacteria-host immune interactions and details the known mechanisms of these interactions to the protein level. A synopsis of the presented membrane surface molecules that elicit particularly promising oncolytic capacities is paired with the stimulated localized and systemic immunogenic effects. In addition, oncolytic bacterial progression toward clinical translation through engineering efforts are discussed, with thorough attention given to strains that have accomplished Phase III clinical trial initiation. In addition to therapeutic mitigation after the tumor has formed, some bacterial species, referred to as "prophylactic", may even be able to prevent or "derail" tumor formation through anti-inflammatory capabilities. These promising species and their particularly favorable characteristics are summarized as well. A complete understanding of the bacteria-host interaction will likely be necessary to assess anti-cancer capacities and unlock the full cancer therapeutic potential of oncolytic bacteria.
Collapse
Affiliation(s)
- Jason M Roe
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Kevin Seely
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Caleb J Bussard
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80130, USA
| | | | - Elizabeth G Mouw
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michael A Hollingsworth
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Amanda E Brooks
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80130, USA
- Office of Research & Scholarly Activity, Rocky Vista University, Ivins, UT 84738, USA
| | - Kaitlin M Dailey
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
3
|
He M, Jiang D, Xun A, Yang J, Luo Q, Wu H. METTL3 enhances PNN mRNA stability through m6A modification to augment tumorigenesis of colon adenocarcinoma. Exp Physiol 2022; 107:1283-1297. [PMID: 35996844 DOI: 10.1113/ep090273] [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: 12/13/2021] [Accepted: 08/12/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? This study focuses on the role of Pinin (PNN) in the malignant phenotype of colon adenocarcinoma cells and the underlying mechanism. What is the main finding and its importance? PNN can be stabilized and upregulated by METTL3, which promotes glycolysis in COAD and augments cell proliferation, migration and invasiveness. METTL3 and PNN might serve as potential targets for the treatment of COAD. ABSTRACT Colon adenocarcinoma (COAD) is a fatal malignancy with high morbidity and mortality rates globally. Pinin (PNN), a desmosome associated protein, has been revealed as a tumor driver in several malignancies. This study aims to probe the expression and role of PNN in COAD and the underlying mechanism. PNN expressed at high levels in clinically collected COAD tumors and was linked to poor prognosis of patients. Downregulation of PNN reduced glucose uptake, lactate production and ATP levels in COAD cells and suppressed cell proliferation, migration, and invasiveness. Methyltransferase like 3 (METTL3) was positively associated with PNN levels in COAD tumor tissues. The RNA immunoprecipitation and m6A quantification assays indicated that METTL3 enhanced PNN mRNA stability and expression in COAD through m6A modification with the involvement of m6A "reader" protein YTHDF1. Downregulation of METTL3 reduced COAD cell glycolysis and proliferation in vitro and suppressed growth and metastasis of xenograft tumors in vivo, but further overexpression of PNN restored malignant behaviors of COAD cells and tumor growth. In summary, this study demonstrates that METTL3 promotes PNN mRNA stability and expression in COAD through m6A modification, which augments glycolysis and proliferation of COAD cells and leads to the resultant tumor progression. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Min He
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, P.R. China
| | - Danling Jiang
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, P.R. China
| | - Anying Xun
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, P.R. China
| | - Jian Yang
- Department of Gastroenterology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, 518067, P.R. China
| | - Qianjiang Luo
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, P.R. China
| | - Huihua Wu
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, P.R. China
| |
Collapse
|
4
|
Yuanchun L, Wenping Z, Jing Z, Wenjie L, Yanlin Z, Yanming L, Jiuxin Q. Mycobacterium paragordonae is an emerging pathogen in human pulmonary disease: clinical features, antimicrobial susceptibility testing and outcomes. Emerg Microbes Infect 2022; 11:1973-1981. [PMID: 35916253 PMCID: PMC9364734 DOI: 10.1080/22221751.2022.2103453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Objectives: Mycobacterium paragordonae (MPG) is an emerging and less common type of Non-tuberculous mycobacteria (NTM) and we know little about its characteristics and prognosis, hence we constructed this retrospective cohort study. Methods: MPG was identified using MALD-TOF MS, multi-target combined gene sequencing and WGS. Clinical information was collected, antimicrobial susceptibility testing was measured using the SLOMYCO panel, and optimal growth temperature testing was measured using Lowenstein-Jensen medium. Results: Eight MPGs were isolated from 1730 NTMs (0.46%); the mean age of MPG pulmonary disease (MPG-PD) patients was 42.38 ± 9.92 years, 37.5% were male, and the average BMI was 18.4 ± 0.51 kg/m2. All patients had the symptoms of cough and sputum and CT images mainly presented in patchy or streaky shadows, MPG grew at 25°C, 30°C and 37°C, and the optimal growth temperature is 37°C. MPGs were sensitive to clarithromycin, rifabutin, amikacin, linezolid, moxifloxacin, cotrimoxazole and ciprofloxacin, two isolates were resistant to rifampicin. Two patients had follow up information, their imaging remained stable during the follow-up. Conclusions: MPG-PD is a rare NTM disease and is more likely to develop in middle-aged, female, and low BMI patients. The patients present with no specific features within the symptoms as well as the CT imaging. The optimal growth temperature of MPG is at 37°C, MPG-PD has excellent sensitivity to drugs recommended by CLSI and presents with a stable disease.
Collapse
Affiliation(s)
- Li Yuanchun
- Department of Clinical Laboratory, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Infectious Diseases (Tuberculosis), Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| | - Zhang Wenping
- Department of Clinical Laboratory, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Infectious Diseases (Tuberculosis), Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| | - Zhao Jing
- Department of Medical Imaging, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Infectious Diseases (Tuberculosis), Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| | - Lai Wenjie
- Department of Clinical Laboratory, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Infectious Diseases (Tuberculosis), Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| | - Zhao Yanlin
- National Tuberculosis Reference Laboratory, Chinese Center for Disease Control and Prevention. Beijing, P.R. China
| | - Li Yanming
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Qu Jiuxin
- Department of Clinical Laboratory, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Infectious Diseases (Tuberculosis), Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| |
Collapse
|
5
|
Jeong H, Lee SY, Seo H, Kim DH, Lee D, Kim BJ. Potential of Mycobacterium tuberculosis chorismate mutase (Rv1885c) as a novel TLR4-mediated adjuvant for dendritic cell-based cancer immunotherapy. Oncoimmunology 2022; 11:2023340. [PMID: 35083095 PMCID: PMC8786331 DOI: 10.1080/2162402x.2021.2023340] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
For clinical application by dendritic cell (DC)-based cancer immunotherapy, a proper adjuvant system to elicit a strong anticancer immune response is needed. Here, we investigated the potential of chorismate mutase (TBCM, Rv1885c), a putative Mycobacterium tuberculosis (TB) virulence factor, as an immunoadjuvant in DC-based tumor immunotherapy. First, we found that TBCM functionally activated DCs by upregulating costimulatory molecules, increasing the secretion of proinflammatory cytokines, enhancing migration and inducing the Th1-type immune response in a dose-dependent manner via TLR4-mediated signaling. In addition, subcutaneous injection of TBCM-activated DCs loaded with cell lysates led to reduced tumor mass, enhanced mouse survival and lowered tumor incidence in lung carcinoma (LLC) cell-bearing mice. This is mainly mediated by functional cytotoxic T lymphocyte-mediated oncolytic activity and inhibition of cancer proliferation- and metastasis-related genes. Moreover, TBCM-induced DCs can also generate memory CD4 T cells and exert long-term tumor prevention effects. In conclusion, our findings suggest that TBCM (Rv1885c), a novel TLR4 agonist, could be used as an immunoadjuvant for DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hyein Jeong
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea
| | - So-Young Lee
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea
- BK21 Four Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyejun Seo
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea
- BK21 Four Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Hyun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea
| | - Duhyung Lee
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea
| |
Collapse
|
6
|
Tan YZ, Yuan T, Tan L, Tian YQ, Long YZ. Lumbar infection caused by Mycobacterium paragordonae: A case report. World J Clin Cases 2021; 9:8879-8887. [PMID: 34734070 PMCID: PMC8546838 DOI: 10.12998/wjcc.v9.i29.8879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/30/2021] [Accepted: 08/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mycobacterium paragordonae (M. paragordonae), a slow-growing, acid-resistant mycobacterial species, was first isolated from the sputum of a lung infection patient in South Korea in 2014. Infections caused by M. paragordonae are rare. CASE SUMMARY Herein, we report the case of a 53-year-old patient who presented with fever and low back pain. Lumbar nuclear magnetic resonance imaging revealed the destruction of the lumbar vertebra with peripheral abscess formation. After anti-infective and diagnostic anti-tuberculosis treatment, the patient had no further fever, but the back pain was not relieved. Postoperatively, the necrotic material was sent for pathological examination, and all tests related to tuberculosis were negative, but pus culture suggested nontuberculous mycobacteria. The necrotic tissue specimens were subjected to metagenomic next-generation sequencing, which indicated the presence of M. paragordonae. Finally, the infecting pathogen was identified, and the treatment plan was adjusted. The patient was in good condition during the follow-up period. CONCLUSION M. paragordonae, a rare nontuberculous mycobacterium, can also cause spinal infections. In the clinic, it is necessary to identify nontuberculous mycobacteria for spinal infections similar to Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Ying-Zheng Tan
- Department of Infectious Diseases, Zhuzhou Central Hospital, Zhuzhou 412000, Hunan Province, China
| | - Ting Yuan
- Department of Infectious Diseases, Zhuzhou Central Hospital, Zhuzhou 412000, Hunan Province, China
| | - Liang Tan
- Department of Spinal Surgery, Zhuzhou Central Hospital, Zhuzhou 412000, Hunan Province, China
| | - Yu-Qiu Tian
- Department of Infectious Diseases, Zhuzhou Central Hospital, Zhuzhou 412000, Hunan Province, China
| | - Yun-Zhu Long
- Department of Infectious Diseases, Zhuzhou Central Hospital, Zhuzhou 412000, Hunan Province, China
| |
Collapse
|
7
|
Carroll CSE, Andrew ER, Malik L, Elliott KF, Brennan M, Meyer J, Hintze A, Almonte AA, Lappin C, MacPherson P, Schulte KM, Dahlstrom JE, Tamhane R, Neeman T, Herbert EW, Orange M, Yip D, Allavena R, Fahrer AM. Simple and effective bacterial-based intratumoral cancer immunotherapy. J Immunother Cancer 2021; 9:jitc-2021-002688. [PMID: 34531247 PMCID: PMC8449973 DOI: 10.1136/jitc-2021-002688] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND We describe intratumoral injection of a slow-release emulsion of killed mycobacteria (complete Freund's adjuvant (CFA)) in three preclinical species and in human cancer patients. METHODS Efficacy and safety were tested in mammary tumors in mice, in mastocytomas in mice and dogs, and in equine melanomas. In mice, survival, tumor growth, and tumor infiltration by six immune cell subsets (by flow cytometry) were investigated and analyzed using Cox proportional hazards, a random slopes model, and a full factorial model, respectively. Tumor growth and histology were investigated in dogs and horses, as well as survival and tumor immunohistochemistry in dogs. Tumor biopsies were taken from human cancer patients on day 5 (all patients) and day 28 (some patients) of treatment and analyzed by histology. CT scans are provided from one patient. RESULTS Significantly extended survival was observed in mouse P815 and 4T1 tumor models. Complete tumor regressions were observed in all three non-human species (6/186 (3%) of mouse mastocytomas; 3/14 (21%) of canine mastocytomas and 2/11 (18%) of equine melanomas). Evidence of systemic immune responses (regression of non-injected metastases) was also observed. Analysis of immune cells infiltrating mastocytoma tumors in mice showed that early neutrophil infiltration was predictive of treatment benefit. Analysis of the site of mastocytoma regression in dogs weeks or months after treatment demonstrated increased B and T cell infiltrates. Thus, activation of the innate immune system alone may be sufficient for regression of some injected tumors, followed by activation of the acquired immune system which can mediate regression of non-injected metastases. Finally, we report on the use of CFA in 12 human cancer patients. Treatment was well tolerated. CT scans showing tumor regression in a patient with late-stage renal cancer are provided. CONCLUSION Our data demonstrate that intratumoral injection of CFA has major antitumor effects in a proportion of treated animals and is safe for use in human cancer patients. Further trials in human cancer patients are therefore warranted. Our novel treatment provides a simple and inexpensive cancer immunotherapy, immediately applicable to a wide range of solid tumors, and is suitable to patients in developing countries and advanced care settings.
Collapse
Affiliation(s)
- Christina S E Carroll
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Erin R Andrew
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Laeeq Malik
- Department of Medical Oncology, Canberra Hospital, Canberra, Australian Capital Territory, Australia.,Medical School, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kathryn F Elliott
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Moira Brennan
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - James Meyer
- Adelaide Plains Equine Clinic, Gawler, South Australia, Australia
| | | | - Andrew A Almonte
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Cassandra Lappin
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Philip MacPherson
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Klaus-Martin Schulte
- Medical School, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jane E Dahlstrom
- Medical School, Australian National University, Canberra, Australian Capital Territory, Australia.,ACT Pathology, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Rohit Tamhane
- Canberra Imaging Group, Canberra, Australian Capital Territory, Australia
| | - Teresa Neeman
- Biological Data Science Institute, Australian National University, Canberra, Australian Capital Territory, Australia
| | | | | | - Desmond Yip
- Department of Medical Oncology, Canberra Hospital, Canberra, Australian Capital Territory, Australia.,Medical School, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Rachel Allavena
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Aude M Fahrer
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
8
|
Kim BJ, Jeong H, Seo H, Lee MH, Shin HM, Kim BJ. Recombinant Mycobacterium paragordonae Expressing SARS-CoV-2 Receptor-Binding Domain as a Vaccine Candidate Against SARS-CoV-2 Infections. Front Immunol 2021; 12:712274. [PMID: 34512635 PMCID: PMC8432291 DOI: 10.3389/fimmu.2021.712274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/04/2021] [Indexed: 01/14/2023] Open
Abstract
At present, concerns that the recent global emergence of SARS-CoV-2 variants could compromise the current vaccines have been raised, highlighting the urgent demand for new vaccines capable of eliciting T cell-mediated immune responses, as well as B cell-mediated neutralizing antibody production. In this study, we developed a novel recombinant Mycobacterium paragordonae expressing the SARS-CoV-2 receptor-binding domain (RBD) (rMpg-RBD-7) that is capable of eliciting RBD-specific immune responses in vaccinated mice. The potential use of rMpg-RBD-7 as a vaccine for SARS-CoV-2 infections was evaluated in in vivo using mouse models of two different modules, one for single-dose vaccination and the other for two-dose vaccination. In a single-dose vaccination model, we found that rMpg-RBD-7 versus a heat-killed strain could exert an enhanced cell-mediated immune (CMI) response, as well as a humoral immune response capable of neutralizing the RBD and ACE2 interaction. In a two-dose vaccination model, rMpg-RBD-7 in a two-dose vaccination could also exert a stronger CMI and humoral immune response to neutralize SARS-CoV-2 infections in pseudoviral or live virus infection systems, compared to single dose vaccinations of rMpg-RBD or two-dose RBD protein immunization. In conclusion, our data showed that rMpg-RBD-7 can lead to an enhanced CMI response and humoral immune responses in mice vaccinated with both single- or two-dose vaccination, highlighting its feasibility as a novel vaccine candidate for SARS-CoV-2. To the best of our knowledge, this study is the first in which mycobacteria is used as a delivery system for a SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Byoung-Jun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Hyein Jeong
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea.,Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea.,BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyejun Seo
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea.,Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea.,Interdisciplinary Program in Cancer Biology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Mi-Hyun Lee
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea.,Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea.,BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun Mu Shin
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea.,Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea.,BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea.,Liver Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea.,Seoul National University Medical Research Center (SNUMRC), Seoul, South Korea.,BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
A Hepatitis B Virus-Derived Peptide Exerts an Anticancer Effect via TNF/iNOS-producing Dendritic Cells in Tumor-Bearing Mouse Model. Cancers (Basel) 2021; 13:cancers13030407. [PMID: 33499256 PMCID: PMC7865762 DOI: 10.3390/cancers13030407] [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: 12/07/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Recently, we reported a 6-mer hepatitis B virus (HBV)-derived peptide, Poly6, that exerts antiviral effects against human immunodeficiency virus type 1 (HIV-1). Here, we explored the immunotherapeutic potential of Poly6 via its administration into dendritic cells (DCs) in a mouse model. Our data revealed that Poly6 treatment led to enhanced production of tumor necrosis factor alpha (TNF-α) and inducible nitric oxide synthase (iNOS)-producing DCs (Tip-DCs) in a type 1 interferon (IFN-I)-dependent manner via the induction of mitochondrial stress. Poly6 treatment in mice implanted with MC38 cells, a murine colon adenocarcinoma line, led to attenuated tumor formation, primarily due to direct cell death induced by Tip-DC mediated nitric oxide (NO) production and indirect killing by Tip-DC mediated cluster of differentiation 8 (CD8) cytotoxic T lymphocyte (CTL) activation via CD40 activation. Moreover, Poly6 treatment demonstrated an enhanced anticancer effect with one of the checkpoint inhibitors, the anti PD-L1 antibody. In conclusion, our data reveal that Poly6 treatment elicits an antitumor immune response in mice, possibly through NO-mediated oncolytic activity via Tip-DC activation and Tip-DC mediated CTL activation. This suggests that Poly6 represents a potential adjuvant for cancer immunotherapy by enhancing the anticancer effects of immune checkpoint inhibitors.
Collapse
|
10
|
Yoon HY, Yang HM, Kim CH, Goo YT, Kang MJ, Lee S, Choi YW. Current status of the development of intravesical drug delivery systems for the treatment of bladder cancer. Expert Opin Drug Deliv 2020; 17:1555-1572. [DOI: 10.1080/17425247.2020.1810016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ho Yub Yoon
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Hee Mang Yang
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | | | - Yoon Tae Goo
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | | | - Sangkil Lee
- College of Pharmacy, Keimyung University, Daegu, Korea
| | | |
Collapse
|
11
|
Mycobacteria-Based Vaccines as Immunotherapy for Non-urological Cancers. Cancers (Basel) 2020; 12:cancers12071802. [PMID: 32635668 PMCID: PMC7408281 DOI: 10.3390/cancers12071802] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
The arsenal against different types of cancers has increased impressively in the last decade. The detailed knowledge of the tumor microenvironment enables it to be manipulated in order to help the immune system fight against tumor cells by using specific checkpoint inhibitors, cell-based treatments, targeted antibodies, and immune stimulants. In fact, it is widely known that the first immunotherapeutic tools as immune stimulants for cancer treatment were bacteria and still are; specifically, the use of Mycobacterium bovis bacillus Calmette-Guérin (BCG) continues to be the treatment of choice for preventing cancer recurrence and progression in non-invasive bladder cancer. BCG and also other mycobacteria or their components are currently under study for the immunotherapeutic treatment of different malignancies. This review focuses on the preclinical and clinical assays using mycobacteria to treat non-urological cancers, providing a wide knowledge of the beneficial applications of these microorganisms to manipulate the tumor microenvironment aiming at tumor clearance.
Collapse
|