1
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
2
|
Singh M, Patel B, Seo M, Ahn P, Wais N, Shen H, Nakka S, Kishore P, Venketaraman V. TB and HIV induced immunosenescence: where do vaccines play a role? FRONTIERS IN AGING 2024; 5:1385963. [PMID: 38903242 PMCID: PMC11188299 DOI: 10.3389/fragi.2024.1385963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/13/2024] [Indexed: 06/22/2024]
Abstract
This paper tackles the complex interplay between Human Immunodeficiency virus (HIV-1) and Mycobacterium tuberculosis (M. tuberculosis) infections, particularly their contribution to immunosenescence, the age-related decline in immune function. Using the current literature, we discuss the immunological mechanisms behind TB and HIV-induced immunosenescence and critically evaluate the BCG (Bacillus Calmette-Guérin) vaccine's role. Both HIV-1 and M. tuberculosis demonstrably accelerate immunosenescence: M. tuberculosis through DNA modification and heightened inflammation, and HIV-1 through chronic immune activation and T cell production compromise. HIV-1 and M. tuberculosis co-infection further hastens immunosenescence by affecting T cell differentiation, underscoring the need for prevention and treatment. Furthermore, the use of the BCG tuberculosis vaccine is contraindicated in patients who are HIV positive and there is a lack of investigation regarding the use of this vaccine in patients who develop HIV co-infection with possible immunosenescence. As HIV does not currently have a vaccine, we focus our review more so on the BCG vaccine response as a result of immunosenescence. We found that there are overall limitations with the BCG vaccine, one of which is that it cannot necessarily prevent re-occurrence of infection due to effects of immunosenescence or protect the elderly due to this reason. Overall, there is conflicting evidence to show the vaccine's usage due to factors involving its production and administration. Further research into developing a vaccine for HIV and improving the BCG vaccine is warranted to expand scientific understanding for public health and beyond.
Collapse
Affiliation(s)
- Mona Singh
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Bhumika Patel
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Michael Seo
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Phillip Ahn
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Nejma Wais
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Haley Shen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - SriHarsha Nakka
- Kempegowda Institute of Medical Sciences, Bengaluru, Karnataka, India
- Masters of Public Health, Chamberlain University, Addison, IL, United States
| | - Priya Kishore
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
3
|
Ozeki Y, Yokoyama A, Nishiyama A, Yoshida Y, Ohara Y, Mashima T, Tomiyama C, Shaban AK, Takeishi A, Osada-Oka M, Yamaguchi T, Tateishi Y, Maeyama JI, Hakamata M, Moro H, Kikuchi T, Hayashi D, Suzuki F, Yamamoto T, Iho S, Katahira M, Yamamoto S, Matsumoto S. Recombinant mycobacterial DNA-binding protein 1 with post-translational modifications boosts IFN-gamma production from BCG-vaccinated individuals' blood cells in combination with CpG-DNA. Sci Rep 2024; 14:9141. [PMID: 38644371 PMCID: PMC11033290 DOI: 10.1038/s41598-024-58836-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/03/2024] [Indexed: 04/23/2024] Open
Abstract
Tuberculosis remains a large health threat, despite the availability of the tuberculosis vaccine, BCG. As BCG efficacy gradually decreases from adolescence, BCG-Prime and antigen-booster may be an efficient strategy to confer vaccine efficacy. Mycobacterial DNA-binding protein 1 (MDP1, namely Rv2986c, hupB or HU) is a major Mycobacterium tuberculosis protein that induces vaccine-efficacy by co-administration with CpG DNA. To produce MDP1 for booster-vaccine use, we have created recombinant MDP1 produced in both Escherichia coli (eMDP1) and Mycolicibacterium smegmatis (mMDP1), an avirulent rapid-growing mycobacteria. We tested their immunogenicity by checking interferon (IFN)-gamma production by stimulated peripheral blood cells derived from BCG-vaccinated individuals. Similar to native M. tuberculosis MDP1, we observed that most lysin resides in the C-terminal half of mMDP1 are highly methylated. In contrast, eMDP1 had less post-translational modifications and IFN-gamma stimulation. mMDP1 stimulated the highest amount of IFN-gamma production among the examined native M. tuberculosis proteins including immunodominant MPT32 and Antigen 85 complex. MDP1-mediated IFN-gamma production was more strongly enhanced when combined with a new type of CpG DNA G9.1 than any other tested CpG DNAs. Taken together, these results suggest that the combination of mMDP1 and G9.1 possess high potential use for human booster vaccine against tuberculosis.
Collapse
Affiliation(s)
- Yuriko Ozeki
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan.
| | - Akira Yokoyama
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-Ku, Tokyo, 113-8654, Japan
| | - Akihito Nishiyama
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Yutaka Yoshida
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Yukiko Ohara
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Tsukasa Mashima
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Chikako Tomiyama
- Graduate School of Health Sciences, Niigata University, 2-746, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8518, Japan
| | - Amina K Shaban
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Atsuki Takeishi
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Mayuko Osada-Oka
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5, Shimogamo-Nakaragi-Cho, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology 1, National Institute of Infectious Disease, 1-23-1, Sinjuku-Ku, Tokyo, 162-8640, Japan
| | - Yoshitaka Tateishi
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Jun-Ichi Maeyama
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Reseach Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, 4-7-1, Musashimurayama, Tokyo, 208-0011, Japan
| | - Mariko Hakamata
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Department of Respiratory Medicine and Infectious Disease, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Hiroshi Moro
- Department of Respiratory Medicine and Infectious Disease, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Disease, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Daisuke Hayashi
- Central Laboratory, Japan BCG Laboratory, 3-1-5 Matsuyama, Kiyose, Tokyo, 204-0022, Japan
| | - Fumiko Suzuki
- Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-Gun, Fukui, 910-1193, Japan
| | - Toshiko Yamamoto
- Central Laboratory, Japan BCG Laboratory, 3-1-5 Matsuyama, Kiyose, Tokyo, 204-0022, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashi-Murayama, Tokyo, 189-0002, Japan
| | - Sumiko Iho
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-Gun, Fukui, 910-1193, Japan
- Louis Pasteur Center for Medical Research, 103-5 Tanaka Monzen-cho, Sakyo-ku, Kyoto, 606-8225, Japan
| | - Masato Katahira
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Saburo Yamamoto
- Central Laboratory, Japan BCG Laboratory, 3-1-5 Matsuyama, Kiyose, Tokyo, 204-0022, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashi-Murayama, Tokyo, 189-0002, Japan
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan.
- Laboratory of Tuberculosis, Institute of Tropical Disease, Universitas Airlangga, Kampus C JI. Mulyorejo, Surabaya, 60113, Indonesia.
- Division of Research Aids, Hokkaido University Institute for Vaccine Research and Development, Kita 20, Nishi 10, Kita-Ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
4
|
Gyu Choi H, Woong Kwon K, Jae Shin S. Importance of adjuvant selection in tuberculosis vaccine development: Exploring basic mechanisms and clinical implications. Vaccine X 2023; 15:100400. [PMID: 37965276 PMCID: PMC10641539 DOI: 10.1016/j.jvacx.2023.100400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
The global emergency of unexpected pathogens, exemplified by SARS-CoV-2, has emphasized the importance of vaccines in thwarting infection and curtailing the progression of severe disease. The scourge of tuberculosis (TB), emanating from the Mycobacterium tuberculosis (Mtb) complex, has inflicted a more profound toll in terms of mortality and morbidity than any other infectious agents prior to the SARS-CoV-2 pandemic. Despite the existence of Bacillus Calmette-Guérin (BCG), the only licensed vaccine developed a century ago, its efficacy against TB remains unsatisfactory, particularly in preventing pulmonary Mtb infections in adolescents and adults. However, collaborations between academic and industrial entities have led to a renewed impetus in the development of TB vaccines, with numerous candidates, particularly subunit vaccines with specialized adjuvants, exhibiting promising outcomes in recent clinical studies. Adjuvants are crucial in modulating optimal immunological responses, by endowing immune cells with sufficient antigen and immune signals. As exemplified by the COVID-19 vaccine landscape, the interplay between vaccine efficacy and adverse effects is of paramount importance, particularly for the elderly and individuals with underlying ailments such as diabetes and concurrent infections. In this regard, adjuvants hold the key to optimizing vaccine efficacy and safety. This review accentuates the pivotal roles of adjuvants and their underlying mechanisms in the development of TB vaccines. Furthermore, we expound on the prospects for the development of more efficacious adjuvants and their synergistic combinations for individuals in diverse states, such as aging, HIV co-infection, and diabetes, by examining the immunological alterations that arise with aging and comparing them with those observed in younger cohorts.
Collapse
Affiliation(s)
- Han Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
5
|
Kaushal A, Khurana I, Yadav P, Allawadhi P, Banothu AK, Neeradi D, Thalugula S, Barani PJ, Naik RR, Navik U, Bharani KK, Khurana A. Advances in therapeutic applications of silver nanoparticles. Chem Biol Interact 2023; 382:110590. [PMID: 37268200 DOI: 10.1016/j.cbi.2023.110590] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
Nanotechnology is one of the most appealing area for developing new applications in biotechnology and medicine. For decades, nanoparticles have been extensively studied for a variety of biomedical applications. Silver has evolved into a potent antibacterial agent that can be used in a variety of nanostructured materials of various shapes and sizes. Silver nanoparticles (AgNP) based antimicrobial compounds are employed in a wide range of applications, including medicinal uses, surface treatment and coatings, the chemical and food industries, and agricultural productivity. When designing formulations for specific applications, the size, shape, and surface area of AgNPs are all crucial structural aspects to consider. Different methods for producing AgNPs with varying sizes and forms that are less harmful have been devised. The anticancer, anti-inflammatory, antibacterial, antiviral, and anti-angiogenic properties of AgNPs have been addressed in this review, as well as their generation and processes. Herein, we have reviewed the advances in therapeutic applications of AgNPs, as well as their limitations and barriers for future applications.
Collapse
Affiliation(s)
- Ashutosh Kaushal
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Isha Khurana
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Prince Allawadhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Anil Kumar Banothu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India
| | - Dinesh Neeradi
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India
| | - Sunitha Thalugula
- Department of Pharmacology, University College of Pharmaceutical Sciences (UCPS), Kakatiya University, Warangal, 506009, Telangana, India
| | - Percy Jasmine Barani
- Department of Chemistry, Wesley Degree College for Women, Osmania University, Secunderabad, 500025, Telangana, India
| | | | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Warangal, 506166, PVNRTVU, Telangana, India.
| | - Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India; Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Warangal, 506166, PVNRTVU, Telangana, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074, Aachen, Germany.
| |
Collapse
|
6
|
Zhuang L, Ye Z, Li L, Yang L, Gong W. Next-Generation TB Vaccines: Progress, Challenges, and Prospects. Vaccines (Basel) 2023; 11:1304. [PMID: 37631874 PMCID: PMC10457792 DOI: 10.3390/vaccines11081304] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), is a prevalent global infectious disease and a leading cause of mortality worldwide. Currently, the only available vaccine for TB prevention is Bacillus Calmette-Guérin (BCG). However, BCG demonstrates limited efficacy, particularly in adults. Efforts to develop effective TB vaccines have been ongoing for nearly a century. In this review, we have examined the current obstacles in TB vaccine research and emphasized the significance of understanding the interaction mechanism between MTB and hosts in order to provide new avenues for research and establish a solid foundation for the development of novel vaccines. We have also assessed various TB vaccine candidates, including inactivated vaccines, attenuated live vaccines, subunit vaccines, viral vector vaccines, DNA vaccines, and the emerging mRNA vaccines as well as virus-like particle (VLP)-based vaccines, which are currently in preclinical stages or clinical trials. Furthermore, we have discussed the challenges and opportunities associated with developing different types of TB vaccines and outlined future directions for TB vaccine research, aiming to expedite the development of effective vaccines. This comprehensive review offers a summary of the progress made in the field of novel TB vaccines.
Collapse
Affiliation(s)
- Li Zhuang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
- Hebei North University, Zhangjiakou 075000, China
| | - Zhaoyang Ye
- Hebei North University, Zhangjiakou 075000, China
| | - Linsheng Li
- Hebei North University, Zhangjiakou 075000, China
| | - Ling Yang
- Hebei North University, Zhangjiakou 075000, China
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| |
Collapse
|
7
|
Moradi M, Vahedi F, Abbassioun A, Ramezanpour Shahi A, Sholeh M, Taheri‐Anganeh M, Dargahi Z, Ghanavati R, Khatami SH, Movahedpour A. Liposomal delivery system/adjuvant for tuberculosis vaccine. Immun Inflamm Dis 2023; 11:e867. [PMID: 37382263 PMCID: PMC10251763 DOI: 10.1002/iid3.867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 06/30/2023] Open
Abstract
As reported by the World Health Organization, about 10 million individuals were infected with tuberculosis (TB) worldwide. Moreover, approximately 1.5 million people died of TB, of which 214,000 were infected with HIV simultaneously. Due to the high infection rate, the need for effective TB vaccination is highly felt. Until now, various methodologies have been proposed for the development of a protein subunit vaccine for TB. These vaccines have shown higher protection than other vaccines, particularly the Bacillus culture vaccine. The delivery system and safety regulator are common characteristics of effective adjuvants in TB vaccines and the clinical trial stage. The present study investigates the current state of TB adjuvant research focusing on the liposomal adjuvant system. Based on our findings, the liposomal system is a safe and efficient adjuvant from nanosize to microsize for vaccinations against TB, other intracellular infections, and malignancies. Clinical studies can provide valuable feedback for developing novel TB adjuvants, which ultimately enhance the impact of adjuvants on next-generation TB vaccines.
Collapse
Affiliation(s)
- Melika Moradi
- Department of Microbiology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Farzaneh Vahedi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Arian Abbassioun
- Department of Virology, Faculty of Veterinary MediceneUniversity of TehranTehranIran
| | - Arash Ramezanpour Shahi
- Department of Veterinary Clinical Sciences, Poultry diseases and hygiene Resident, Faculty of Veterinary MedicineShahrekord UniversityShahrekordIran
| | - Mohammad Sholeh
- Department of BacteriologyPasteur Institute of IranTehranIran
| | - Mortaza Taheri‐Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research InstituteUrmia University of Medical SciencesUrmiaIran
| | - Zahra Dargahi
- Department of Microbiology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | | | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
8
|
Kwon KW, Kang TG, Lee A, Jin SM, Lim YT, Shin SJ, Ha SJ. Protective Efficacy and Immunogenicity of Rv0351/Rv3628 Subunit Vaccine Formulated in Different Adjuvants Against Mycobacterium tuberculosis Infection. Immune Netw 2023; 23:e16. [PMID: 37179749 PMCID: PMC10166659 DOI: 10.4110/in.2023.23.e16] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 05/15/2023] Open
Abstract
Bacillus Calmette-Guerin (BCG) vaccine is the only licensed vaccine for tuberculosis (TB) prevention. Previously, our group demonstrated the vaccine potential of Rv0351 and Rv3628 against Mycobacterium tuberculosis (Mtb) infection by directing Th1-biased CD4+ T cells co-expressing IFN-γ, TNF-α, and IL-2 in the lungs. Here, we assessed immunogenicity and vaccine potential of the combined Ags (Rv0351/Rv3628) formulated in different adjuvants as subunit booster in BCG-primed mice against hypervirulent clinical Mtb strain K (Mtb K). Compared to BCG-only or subunit-only vaccine, BCG prime and subunit boost regimen exhibited significantly enhanced Th1 response. Next, we evaluated the immunogenicity to the combined Ags when formulated with four different types of monophosphoryl lipid A (MPL)-based adjuvants: 1) dimethyldioctadecylammonium bromide (DDA), MPL, and trehalose dicorynomycolate (TDM) in liposome form (DMT), 2) MPL and Poly I:C in liposome form (MP), 3) MPL, Poly I:C, and QS21 in liposome form (MPQ), and 4) MPL and Poly I:C in squalene emulsion form (MPS). MPQ and MPS displayed greater adjuvancity in Th1 induction than DMT or MP did. Especially, BCG prime and subunit-MPS boost regimen significantly reduced the bacterial loads and pulmonary inflammation against Mtb K infection when compared to BCG-only vaccine at a chronic stage of TB disease. Collectively, our findings highlighted the importance of adjuvant components and formulation to induce the enhanced protection with an optimal Th1 response.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Tae Gun Kang
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Ara Lee
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
9
|
Duong VT, Skwarczynski M, Toth I. Towards the development of subunit vaccines against tuberculosis: The key role of adjuvant. Tuberculosis (Edinb) 2023; 139:102307. [PMID: 36706503 DOI: 10.1016/j.tube.2023.102307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
According to the World Health Organization (WHO), tuberculosis (TB) is the leading cause of death triggered by a single infectious agent, worldwide. Bacillus Calmette-Guerin (BCG) is the only currently licensed anti-TB vaccine. However, other strategies, including modification of recombinant BCG vaccine, attenuated Mycobacterium tuberculosis (Mtb) mutant constructs, DNA and protein subunit vaccines, are under extensive investigation. As whole pathogen vaccines can trigger serious adverse reactions, most current strategies are focused on the development of safe anti-TB subunit vaccines; this is especially important given the rising TB infection rate in immunocompromised HIV patients. The whole Mtb genome has been mapped and major antigens have been identified; however, optimal vaccine delivery mode is still to be established. Isolated protein antigens are typically poorly immunogenic so adjuvants are required to induce strong and long-lasting immune responses. This article aims to review the developmental status of anti-TB subunit vaccine adjuvants.
Collapse
Affiliation(s)
- Viet Tram Duong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
10
|
Romano M, Squeglia F, Kramarska E, Barra G, Choi HG, Kim HJ, Ruggiero A, Berisio R. A Structural View at Vaccine Development against M. tuberculosis. Cells 2023; 12:317. [PMID: 36672252 PMCID: PMC9857197 DOI: 10.3390/cells12020317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Tuberculosis (TB) is still the leading global cause of death from an infectious bacterial agent. Limiting tuberculosis epidemic spread is therefore an urgent global public health priority. As stated by the WHO, to stop the spread of the disease we need a new vaccine, with better coverage than the current Mycobacterium bovis BCG vaccine. This vaccine was first used in 1921 and, since then, there are still no new licensed tuberculosis vaccines. However, there is extremely active research in the field, with a steep acceleration in the past decades, due to the advance of technologies and more rational vaccine design strategies. This review aims to gather latest updates in vaccine development in the various clinical phases and to underline the contribution of Structural Vaccinology (SV) to the development of safer and effective antigens. In particular, SV and the development of vaccine adjuvants is making the use of subunit vaccines, which are the safest albeit the less antigenic ones, an achievable goal. Indeed, subunit vaccines overcome safety concerns but need to be rationally re-engineered to enhance their immunostimulating effects. The larger availability of antigen structural information as well as a better understanding of the complex host immune response to TB infection is a strong premise for a further acceleration of TB vaccine development.
Collapse
Affiliation(s)
- Maria Romano
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Flavia Squeglia
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Eliza Kramarska
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Giovanni Barra
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Alessia Ruggiero
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| |
Collapse
|
11
|
Maurya P, Saklani R, Singh S, Nisha R, Mishra N, Singh P, Pal RR, Kumar A, Chourasia MK, Saraf SA. Effective uptake of folate-functionalized ethionamide-loaded hybrid system: targeting alveolar macrophages. Nanomedicine (Lond) 2022; 17:1819-1831. [PMID: 36136373 DOI: 10.2217/nnm-2021-0468] [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] [Indexed: 01/10/2023] Open
Abstract
Aim: To assess the targeting ability of hybrid nanosystems functionalized with folate. It also aimed to reduce stomach intolerance by substituting the oral route for parenteral delivery. Method: The nanosystems, prepared by nanoprecipitation technique, utilized a one-step method to prepare nanoparticles followed by surface functionalization through adsorption. The prepared nanosystems underwent physical characterization, in vitro and in vivo evaluations. Result: The nanosystems were effective in targeting the alveolar macrophages. Ethionamide was released from the formulation over 5 days. Fourier-transform infrared results proved the structural characteristics, and the positive charge further improved the targeting efficacy on the functionalized system. Conclusion: The hybrid formulation improved the release characteristics. Reduction in dosing frequency due to prolonged release improves compliance with the dosage regimen.
Collapse
Affiliation(s)
- Priyanka Maurya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| | - Ravi Saklani
- Department of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Lucknow, 226031, India
| | - Samipta Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| | - Raquibun Nisha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| | - Nidhi Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| | - Priya Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| | - Ravi Raj Pal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| | - Abhiram Kumar
- Department of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Lucknow, 226031, India
| | - Manish K Chourasia
- Department of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Lucknow, 226031, India
| | - Shubhini A Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University) Vidya Vihar, Lucknow, 226025, India
| |
Collapse
|
12
|
Synthesis and evaluation of inhibitors of Mycobacterium tuberculosis UGM using bioisosteric replacement. Bioorg Med Chem 2022; 69:116896. [DOI: 10.1016/j.bmc.2022.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/08/2022] [Accepted: 06/20/2022] [Indexed: 11/22/2022]
|
13
|
Saelee C, Hanthamrongwit J, Soe PT, Khaenam P, Inthasin N, Ekpo P, Chootong P, Leepiyasakulchai C. Toll-like receptor-mediated innate immune responses by recognition of the recombinant dormancy-associated Mycobacterium tuberculosis proteins Rv2659c and Rv1738. PLoS One 2022; 17:e0273517. [PMID: 36048884 PMCID: PMC9436120 DOI: 10.1371/journal.pone.0273517] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) poses a major threat to the global public health. Importantly, latent tuberculosis infection (LTBI) still impedes the elimination of TB incidence since it has a substantial risk to develop active disease. A multi-stage subunit vaccine comprising active and latency antigens of Mtb has been raised as the promising vaccine to trigger immune protection against all stages of TB. Therefore, the discovery of new antigens that could trigger broad immune response is essential. While current development of TB vaccine mainly focuses on protective immunity mediated by adaptive immune response, the knowledge on triggering the innate immune response by antigens is still limited. We showed that recombinant dormancy-associated Mtb proteins Rv2659c and Rv1738 were recognized by human innate immune recognition molecules, Toll-like receptors (TLRs) 2 and 4 by using HEK-Blue™ hTLR2/hTLR4 systems. We further demonstrated that these two proteins activated phosphorylated NF-κB p65 (Ser536) in the human CD14+ blood cells. We also investigated that these two proteins significantly induced level of pro- and anti-inflammatory cytokines (IL-1β, IL-6, IL-8, IL-10 and TNF-α) which were mediated through TLR2 and TLR4 pathways in human peripheral blood mononuclear cells (hPBMCs). These findings suggest that proteins Rv2659c and Rv1738 stimulated innate immune response targeting TLR2 and TLR4 to produce inflammatory cytokines, and their benefits would be valuable for the development of an effective prophylactic tuberculosis vaccine.
Collapse
Affiliation(s)
- Chutiphon Saelee
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Jariya Hanthamrongwit
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Phyu Thwe Soe
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
- Department of Medical Laboratory Technology, University of Medical Technology, Mandalay, Myanmar
| | - Prasong Khaenam
- Faculty of Medical Technology, Center of Standardization and Product Validation, Mahidol University, Bangkok, Thailand
| | - Naharuthai Inthasin
- Faculty of Medicine Siriraj Hospital, Department of Immunology, Mahidol University, Bangkok, Thailand
| | - Pattama Ekpo
- Faculty of Medicine Siriraj Hospital, Department of Immunology, Mahidol University, Bangkok, Thailand
| | - Patchanee Chootong
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Chaniya Leepiyasakulchai
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
14
|
Kadir R, Luwi NM, Ahmad S, Azlyna AN, Nordin A, Sarmiento M, Acosta A, Azmi M, Uskoković V, Mohamud R. Liposomes as immunological adjuvants and delivery systems in the development of tuberculosis vaccine: A review. ASIAN PAC J TROP MED 2022. [DOI: 10.4103/1995-7645.332806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
15
|
Qu M, Zhou X, Li H. BCG vaccination strategies against tuberculosis: updates and perspectives. Hum Vaccin Immunother 2021; 17:5284-5295. [PMID: 34856853 DOI: 10.1080/21645515.2021.2007711] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Bacillus Calmette-Guérin (BCG) is the only licensed vaccine against tuberculosis (TB). However, BCG has variable efficacy and cannot completely prevent TB infection and transmission. Therefore, the worldwide prevalence of TB calls for urgent development of a more effective TB vaccine. In the absence of other approved vaccines, it is also necessary to improve the efficacy of BCG itself. Intravenous (IV) BCG administration and BCG revaccination strategies have recently shown promising results for clinical usage. Therefore, it is necessary for us to revisit the BCG vaccination strategies and summarize the current research updates related to BCG vaccination. This literature review provides an updated overview and perspectives of the immunization strategies against TB using BCG, which may inspire the following research on TB vaccine development.
Collapse
Affiliation(s)
- Mengjin Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, China Agricultural University, Beijing, China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
16
|
Mustafa AS. Adjuvants and Antigen-Delivery Systems for Subunit Vaccines against Tuberculosis. Vaccines (Basel) 2021; 9:vaccines9090972. [PMID: 34579209 PMCID: PMC8472090 DOI: 10.3390/vaccines9090972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
The only licensed vaccine against tuberculosis is BCG. However, BCG has failed to provide consistent protection against tuberculosis, especially pulmonary disease in adults. Furthermore, the use of BCG is contraindicated in immunocompromised subjects. The research towards the development of new vaccines against TB includes the use of Mycobacterium tuberculosis antigens as subunit vaccines. Such vaccines may be used either alone or in the prime-boost model in BCG-vaccinated people. However, the antigens for subunit vaccines require adjuvants and/or delivery systems to induce appropriate and protective immune responses against tuberculosis and other diseases. Articles published in this Special Issue have studied the pathogenesis of BCG in children and the use of BCG and recombinant BCG as potential vaccines against asthma. Furthermore, the use of different adjuvants and delivery systems in inducing the protective immune responses after immunization with subunit vaccines has been described.
Collapse
Affiliation(s)
- Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| |
Collapse
|
17
|
Bellini C, Horváti K. Recent Advances in the Development of Protein- and Peptide-Based Subunit Vaccines against Tuberculosis. Cells 2020; 9:cells9122673. [PMID: 33333744 PMCID: PMC7765234 DOI: 10.3390/cells9122673] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The World Health Organization (WHO) herald of the “End TB Strategy” has defined goals and targets for tuberculosis prevention, care, and control to end the global tuberculosis endemic. The emergence of drug resistance and the relative dreadful consequences in treatment outcome has led to increased awareness on immunization against Mycobacterium tuberculosis (Mtb). However, the proven limited efficacy of Bacillus Calmette-Guérin (BCG), the only licensed vaccine against Mtb, has highlighted the need for alternative vaccines. In this review, we seek to give an overview of Mtb infection and failure of BCG to control it. Afterward, we focus on the protein- and peptide-based subunit vaccine subtype, examining the advantages and drawbacks of using this design approach. Finally, we explore the features of subunit vaccine candidates currently in pre-clinical and clinical evaluation, including the antigen repertoire, the exploited adjuvanted delivery systems, as well as the spawned immune response.
Collapse
Affiliation(s)
- Chiara Bellini
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary;
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Kata Horváti
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary
- Correspondence:
| |
Collapse
|
18
|
Safar HA, Mustafa AS, McHugh TD. COVID-19 vaccine development: What lessons can we learn from TB? Ann Clin Microbiol Antimicrob 2020; 19:56. [PMID: 33256750 PMCID: PMC7702199 DOI: 10.1186/s12941-020-00402-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
At the time of writing, the SARS-CoV-2 virus has infected more than 49 million people causing more than 1.2 million deaths worldwide since its emergence from Wuhan, China in December 2019. Vaccine development against SARS-CoV-2 has drawn the global attention in order to stop the spread of the virus, with more than 10 vaccines being tested in phase III clinical trials, as of November 2020. However, critical to vaccine development is consideration of the immunological response elicited as well as biological features of the vaccine and both need to be evaluated thoroughly. Tuberculosis is also a major infectious respiratory disease of worldwide prevalence and the vaccine development for tuberculosis has been ongoing for decades. In this review, we highlight some of the common features, challenges and complications in tuberculosis vaccine development, which may also be relevant for, and inform, COVID-19 vaccine development.
Collapse
Affiliation(s)
- Hussain A Safar
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK.
| | - Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Timothy D McHugh
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
19
|
Abstract
Mycobacterium tuberculosis remains the leading cause of death attributed to a single infectious organism. Bacillus Calmette-Guerin (BCG), the standard vaccine against M. tuberculosis, is thought to prevent only 5% of all vaccine-preventable deaths due to tuberculosis, thus an alternative vaccine is required. One of the principal barriers to vaccine development against M. tuberculosis is the complexity of the immune response to infection, with uncertainty as to what constitutes an immunological correlate of protection. In this paper, we seek to give an overview of the immunology of M. tuberculosis infection, and by doing so, investigate possible targets of vaccine development. This encompasses the innate, adaptive, mucosal and humoral immune systems. Though MVA85A did not improve protection compared with BCG alone in a large-scale clinical trial, the correlates of protection this has revealed, in addition to promising results from candidate such as VPM1002, M72/ASO1E and H56:IC31 point to a brighter future in the field of TB vaccine development.
Collapse
Affiliation(s)
- Benedict Brazier
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| |
Collapse
|
20
|
Investigating the Impact of Delivery System Design on the Efficacy of Self-Amplifying RNA Vaccines. Vaccines (Basel) 2020; 8:vaccines8020212. [PMID: 32397231 PMCID: PMC7348957 DOI: 10.3390/vaccines8020212] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022] Open
Abstract
messenger RNA (mRNA)-based vaccines combine the positive attributes of both live-attenuated and subunit vaccines. In order for these to be applied for clinical use, they require to be formulated with delivery systems. However, there are limited in vivo studies which compare different delivery platforms. Therefore, we have compared four different cationic platforms: (1) liposomes, (2) solid lipid nanoparticles (SLNs), (3) polymeric nanoparticles (NPs) and (4) emulsions, to deliver a self-amplifying mRNA (SAM) vaccine. All formulations contained either the non-ionizable cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) or dimethyldioctadecylammonium bromide (DDA) and they were characterized in terms of physico-chemical attributes, in vitro transfection efficiency and in vivo vaccine potency. Our results showed that SAM encapsulating DOTAP polymeric nanoparticles, DOTAP liposomes and DDA liposomes induced the highest antigen expression in vitro and, from these, DOTAP polymeric nanoparticles were the most potent in triggering humoral and cellular immunity among candidates in vivo.
Collapse
|
21
|
Kim H, Kwon KW, Park J, Kang H, Lee Y, Sohn EJ, Hwang I, Eum SY, Shin SJ. Plant-Produced N-glycosylated Ag85A Exhibits Enhanced Vaccine Efficacy Against Mycobacterium tuberculosis HN878 Through Balanced Multifunctional Th1 T Cell Immunity. Vaccines (Basel) 2020; 8:vaccines8020189. [PMID: 32325740 PMCID: PMC7349862 DOI: 10.3390/vaccines8020189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 11/16/2022] Open
Abstract
Tuberculosis (TB) is one of the deadliest infectious diseases worldwide and is caused by Mycobacterium tuberculosis (Mtb). An effective vaccine to prevent TB is considered the most cost-effective measure for controlling this disease. Many different vaccine antigen (Ag) candidates, including well-known and newly identified Ags, have been evaluated in clinical and preclinical studies. In this study, we took advantage of a plant system of protein expression using Nicotiana benthamiana to produce N-glycosylated antigen 85A (G-Ag85A), which is one of the most well-characterized vaccine Ag candidates in the field of TB vaccines, and compared its immunogenicity and vaccine efficacy with those of nonglycosylated Ag85A (NG-Ag85A) produced with an Escherichia coli system. Notably, G-Ag85A induced a more robust IFN-γ response than NG-Ag85A, which indicated that G-Ag85A is well recognized by the host immune system during Mtb infection. We subsequently compared the vaccine potential of G-Ag85A and NG-Ag85A by evaluating their immunological features and substantial protection efficacies. Interestingly, G-Ag85A yielded moderately enhanced long-term protective efficacy, as measured in terms of bacterial burden and lung inflammation. Strikingly, G-Ag85A-immunized mice showed a more balanced proportion of multifunctional Th1-biased immune responses with sustained IFN-γ response than did NG-Ag85A-immunized mice. Collectively, plant-derived G-Ag85A could induce protective and balanced Th1 responses and confer long-term protection against a hypervirulent Mtb Beijing strain infection, which indicated that plant-produced G-Ag85A might provide an excellent example for the production of an Mtb subunit vaccine Ag and could be an effective platform for the development of anti-TB vaccines.
Collapse
Affiliation(s)
- Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
| | - Jaehun Park
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
| | - Hyangju Kang
- BioApplications Inc., Pohang 37668, Korea; (H.K.); (Y.L.); (E.-J.S.)
| | - Yongjik Lee
- BioApplications Inc., Pohang 37668, Korea; (H.K.); (Y.L.); (E.-J.S.)
| | - Eun-Ju Sohn
- BioApplications Inc., Pohang 37668, Korea; (H.K.); (Y.L.); (E.-J.S.)
- School of Interdisciplinary Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Inhwan Hwang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea;
| | - Seok-Yong Eum
- Division of Immunopathology and Cellular Immunology, International Tuberculosis Research Center, Changwon 51755, Korea;
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
- Correspondence:
| |
Collapse
|
22
|
Garcia-Vilanova A, Chan J, Torrelles JB. Underestimated Manipulative Roles of Mycobacterium tuberculosis Cell Envelope Glycolipids During Infection. Front Immunol 2019; 10:2909. [PMID: 31921168 PMCID: PMC6930167 DOI: 10.3389/fimmu.2019.02909] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
The Mycobacterium tuberculosis cell envelope has been evolving over time to make the bacterium transmissible and adaptable to the human host. In this context, the M. tuberculosis cell envelope contains a peripheral barrier full of lipids, some of them unique, which confer M. tuberculosis with a unique shield against the different host environments that the bacterium will encounter at the different stages of infection. This lipid barrier is mainly composed of glycolipids that can be characterized by three different subsets: trehalose-containing, mannose-containing, and 6-deoxy-pyranose-containing glycolipids. In this review, we explore the roles of these cell envelope glycolipids in M. tuberculosis virulence and pathogenesis, drug resistance, and further, how these glycolipids may dictate the M. tuberculosis cell envelope evolution from ancient to modern strains. Finally, we address how these M. tuberculosis cell envelope glycolipids are impacted by the host lung alveolar environment, their role in vaccination and masking host immunity, and subsequently the impact of these glycolipids in shaping how M. tuberculosis interacts with host cells, manipulating their immune response to favor the establishment of an infection.
Collapse
Affiliation(s)
- Andreu Garcia-Vilanova
- Population Health Program, TB Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - John Chan
- Department of Medicine (Infectious Diseases), Albert Einstein College of Medicine & Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine & Montefiore Medical Center, Bronx, NY, United States
| | - Jordi B. Torrelles
- Population Health Program, TB Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
23
|
Vasina DV, Kleymenov DA, Manuylov VA, Mazunina EP, Koptev EY, Tukhovskaya EA, Murashev AN, Gintsburg AL, Gushchin VA, Tkachuk AP. First-In-Human Trials of GamTBvac, a Recombinant Subunit Tuberculosis Vaccine Candidate: Safety and Immunogenicity Assessment. Vaccines (Basel) 2019; 7:vaccines7040166. [PMID: 31683812 PMCID: PMC6963980 DOI: 10.3390/vaccines7040166] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis is known to be the biggest global health problem, causing the most deaths by a single infectious agent. Vaccine-development efforts are extremely important. This paper represents the results of the first-in-human trial of recombinant subunit tuberculosis vaccine GamTBvac in a Phase I study. GamTBvac is a new BCG booster candidate vaccine containing dextran-binding domain modified Ag85a and ESAT6-CFP10 MTB antigens and CpG ODN adjuvant, formulated with dextrans. Safety and immunogenicity of GamTBvac were estimated in an open-label clinical trial on 60 Mycobacterium tuberculosis uninfected (MTB-uninfected) volunteers previously-vaccinated with Bacillus Calmette—Guérin vaccine (BCG). The candidate vaccine had an acceptable safety profile and was well-tolerated. Three different vaccine doses with a double-immunization scheme were assessed for immunogenicity and induced a significant increase in IFN-γ in-house IGRA response and IgG ELISA analysis. Among them, the half dose vaccine group (containing DBD-ESAT6-CFP10, 12.5 μg; DBD-Ag85a, 12.5 μg; CpG (ODN 2216), 75 μg; DEAE-Dextran 500 kDa, 250 μg; and Dextran 500 kDa, 5 mg) provided high, early and stable in time immune response specific to both protein antigen fusions and is proposed for the further studies.
Collapse
Affiliation(s)
- Daria V Vasina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Denis A Kleymenov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Victor A Manuylov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena P Mazunina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Egor Yu Koptev
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena A Tukhovskaya
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Arkady N Murashev
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Alexander L Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Infectology Department, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Vladimir A Gushchin
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Artem P Tkachuk
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| |
Collapse
|
24
|
Van Dis E, Sogi KM, Rae CS, Sivick KE, Surh NH, Leong ML, Kanne DB, Metchette K, Leong JJ, Bruml JR, Chen V, Heydari K, Cadieux N, Evans T, McWhirter SM, Dubensky TW, Portnoy DA, Stanley SA. STING-Activating Adjuvants Elicit a Th17 Immune Response and Protect against Mycobacterium tuberculosis Infection. Cell Rep 2019; 23:1435-1447. [PMID: 29719256 DOI: 10.1016/j.celrep.2018.04.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/09/2018] [Accepted: 03/30/2018] [Indexed: 01/04/2023] Open
Abstract
There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen.
Collapse
Affiliation(s)
- Erik Van Dis
- Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kimberly M Sogi
- School of Public Health, Division of Infectious Disease and Vaccinology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chris S Rae
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | - Kelsey E Sivick
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | - Natalie H Surh
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | | | - David B Kanne
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | - Ken Metchette
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | - Justin J Leong
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | - Jacob R Bruml
- Aduro Biotech, Inc., 740 Heinz Avenue, Berkeley, CA 94710, USA
| | - Vivian Chen
- School of Public Health, Division of Infectious Disease and Vaccinology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kartoosh Heydari
- LKS Flow Cytometry Core, Cancer Research Laboratory, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Tom Evans
- Vaccitech Limited, King Charles House, Park End Street, Oxford OX1 1JD, UK
| | | | | | - Daniel A Portnoy
- Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA; School of Public Health, Division of Infectious Disease and Vaccinology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sarah A Stanley
- Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, CA 94720, USA; School of Public Health, Division of Infectious Disease and Vaccinology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
25
|
Adjuvant Strategies for More Effective Tuberculosis Vaccine Immunity. Microorganisms 2019; 7:microorganisms7080255. [PMID: 31409028 PMCID: PMC6724148 DOI: 10.3390/microorganisms7080255] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/03/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis infection is responsible for the most deaths by a single infectious agent worldwide, with 1.6 million deaths in 2017 alone. The World Health Organization, through its "End TB" strategy, aims to reduce TB deaths by 95% by 2035. In order to reach this goal, a more effective vaccine than the Bacillus Calmette-Guerin (BCG) vaccine currently in use is needed. Subunit TB vaccines are ideal candidates, because they can be used as booster vaccinations for individuals who have already received BCG and would also be safer for use in immunocompromised individuals in whom BCG is contraindicated. However, subunit TB vaccines will almost certainly require formulation with a potent adjuvant. As the correlates of vaccine protection against TB are currently unclear, there are a variety of adjuvants currently being used in TB vaccines in preclinical and clinical development. This review describes the various adjuvants in use in TB vaccines, their effectiveness, and their proposed mechanisms of action. Notably, adjuvants with less inflammatory and reactogenic profiles that can be administered safely via mucosal routes, may have the biggest impact on future directions in TB vaccine design.
Collapse
|
26
|
Malik A, Gupta M, Mani R, Bhatnagar R. Single-dose Ag85B-ESAT6-loaded poly(lactic- co-glycolic acid) nanoparticles confer protective immunity against tuberculosis. Int J Nanomedicine 2019; 14:3129-3143. [PMID: 31118627 PMCID: PMC6501725 DOI: 10.2147/ijn.s172391] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Bacillus Calmette-Guérin, the attenuated strain of Mycobacterium bovis, remains the only available vaccine against tuberculosis (TB). However, its ineffectiveness in adults against pulmonary TB and varied protective efficacy (0-80%) speak to an urgent need for the development of an improved and efficient TB vaccine. In this milieu, poly(lactic-co-glycolic acid) (PLGA), is a preferential candidate, due to such properties as biocompatibility, targeted delivery, sustained antigen release, and atoxic by-products. METHODS In this study, we formulated PLGA nanoparticles (NPs) encapsulating the bivalent H1 antigen, a fusion of Mycobacterium tuberculosis (Mtb) Ag85B and ESAT6 proteins, and investigated its role in immunomodulation and protection against Mtb challenge. Using the classical water-oil-water solvent-evaporation method, H1-NPs were prepared, with encapsulation efficiency of 86.1%±3.2%. These spherical NPs were ~244.4±32.6 nm in diameter, with a negatively charged surface (ζ-potential -4±0.6 mV). RESULTS Under physiological conditions, NPs degraded slowly and the encapsulated H1 antigen was released over a period of weeks. As a proof-of-concept vaccine candidate, H1 NPs were efficiently internalized by the THP-1 human macrophages. Six weeks after a single-dose vaccination, H1 NP-immunized C57BL/6J mice showed significant increase in the production of total serum IgG (P<0.0001) and its isotypes compared to H1 alone, IgG2a being the predominant one, followed by IgG1. Further, the cytokine-release profile of antigen-stimulated splenocyteculture supernatant indicated a strong TH1-biased immunoresponse in H1 NP-vaccinated mice, with ~6.03- and ~2.8-fold increase in IFNγ and TNFα cytokine levels, and ~twofold and 1.6 fold increase in IL4 and IL10 cytokines, respectively, compared to H1 alone-immunized mice. In protection studies, H1 NP-vaccinated mice displayed significant reductions in lung and spleen bacillary load (P<0.05) at 5-week post-Mtb H37Rv challenge and prolonged survival, with a mean survival time of 177 days, compared to H1 alone-vaccinated mice (mean survival time 80 days). CONCLUSION Altogether, our findings highlight the significance of the H1-PLGA nanoformulation in terms of providing long-term protection in mice with a single dose.
Collapse
Affiliation(s)
- Anshu Malik
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| | - Manish Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| | - Rajesh Mani
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| |
Collapse
|
27
|
Moreno-Mendieta S, Barrera-Rosales A, Mata-Espinosa D, Barrios-Payán J, Sánchez S, Hernández-Pando R, Rodríguez-Sanoja R. Raw starch microparticles as BCG adjuvant: Their efficacy depends on the virulence of the infection strains. Vaccine 2019; 37:5731-5737. [PMID: 31000412 DOI: 10.1016/j.vaccine.2019.04.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 02/06/2023]
Abstract
The persistence of tuberculosis (TB) as one of the top 10 causes of death worldwide, the growing incidence of multidrug-resistant tuberculosis and the controversial efficacy of the Bacille Calmette-Guérin (BCG) vaccine drives the development of new generation multistage vaccines against this disease that can boost BCG-primed immunity. The use of polymeric microparticles for this purpose increases due to their advantages, especially their good safety levels and intrinsic immunostimulant properties. We recently explored and demonstrated the reinforcing and adjuvant potential of starch microparticles (SMPs) that administered intranasally to BCG-primed BALB/c mice, alone or in combination with a recombinant antigen, increased survival rates and induced a reduction of bacterial load in the lungs of mice infected with tuberculosis. Here, we tested the effect of SMPs added to the BCG vaccine as adjuvant to the whole-cell vaccine and investigated their contribution to the improvement of the protective efficacy of subcutaneous vaccination in mice challenged with virulent strains of Mycobacterium tuberculosis. As expected, our results were dependent on the infection strains, showing that virulence is a crucial factor that affects the adjuvant activity of SMPs. Our results also confirm the adjuvant activity of this carbohydrate and its usefulness in diverse vaccination strategies not only for mucosal but also for parenteral administration.
Collapse
Affiliation(s)
- Silvia Moreno-Mendieta
- CONACYT, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico; Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Alejandra Barrera-Rosales
- Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Dulce Mata-Espinosa
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Jorge Barrios-Payán
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Sergio Sánchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico.
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| |
Collapse
|
28
|
Chesson CB, Huante M, Nusbaum RJ, Walker AG, Clover TM, Chinnaswamy J, Endsley JJ, Rudra JS. Nanoscale Peptide Self-assemblies Boost BCG-primed Cellular Immunity Against Mycobacterium tuberculosis. Sci Rep 2018; 8:12519. [PMID: 30131591 PMCID: PMC6104033 DOI: 10.1038/s41598-018-31089-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/23/2018] [Indexed: 11/30/2022] Open
Abstract
Bacillus Calmette-Guerin (BCG) is the only vaccine against TB and has limited protection efficacy, which wanes past adolescence. Multifunctional CD8+ T cells (IFN-γ+/TNF-α+/IL-2+) are associated with lower reactivation risk and enhanced control of active Mtb infection. Since boosting with BCG is contraindicated, booster vaccines that augment T cell immunity in the lungs of BCG-vaccinated individuals are urgently needed. We developed a vaccination strategy based on self-assembling peptide nanofibers presenting Mtb-specific CD8+ or CD4+ T cell epitopes that induce high frequency and antigen-specific effector memory T cells producing IFN-γ and IL-2. Intranasal immunization with peptide nanofibers was well tolerated in mice leading to increased antigen-specific CD8+ T cell population in the lungs. Co-assembled nanofibers of CD8+ T cell epitopes and toll-like receptor 2 (TLR2) agonists induced a 8-fold expansion in multifunctional CD8+ T cell populations in the lungs of vaccinated mice. Aerosol challenge with Mtb in BCG-primed and nanofiber-boosted mice provided an additional 0.5-log CFU reduction in lung bacterial load and indicating enhanced protection compared to BCG alone. Together, these data suggest that heterologous prime-boost with BCG and peptide nanofiber vaccines induces cell mediated immunity in the lung, reduces bacterial burden, and is a potentially safer alternative for boosting BCG-primed immunity.
Collapse
Affiliation(s)
- Charles B Chesson
- Department of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08823, USA
| | - Matthew Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Rebecca J Nusbaum
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Aida G Walker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, 77555, Texas, USA
| | - Tara M Clover
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, 77555, Texas, USA
| | - Jagannath Chinnaswamy
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Jai S Rudra
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, 77555, Texas, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
29
|
Joslyn LR, Pienaar E, DiFazio RM, Suliman S, Kagina BM, Flynn JL, Scriba TJ, Linderman JJ, Kirschner DE. Integrating Non-human Primate, Human, and Mathematical Studies to Determine the Influence of BCG Timing on H56 Vaccine Outcomes. Front Microbiol 2018; 9:1734. [PMID: 30177914 PMCID: PMC6109686 DOI: 10.3389/fmicb.2018.01734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/11/2018] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death by an infectious agent, and developing an effective vaccine is an important component of the WHO's EndTB Strategy. Non-human primate (NHP) models of vaccination are crucial to TB vaccine development and have informed design of subsequent human trials. However, challenges emerge when translating results from animal models to human applications, and connecting post-vaccination immunological measurements to infection outcomes. The H56:IC31 vaccine is a candidate currently in phase I/IIa trials. H56 is a subunit vaccine that is comprised of 3 mycobacterial antigens: ESAT6, Ag85B, and Rv2660, formulated in IC31 adjuvant. H56, as a boost to Bacillus Calmette-Guérin (BCG, the TB vaccine that is currently used in most countries world-wide) demonstrates improved protection (compared to BCG alone) in mouse and NHP models of TB, and the first human study of H56 reported strong antigen-specific T cell responses to the vaccine. We integrated NHP and human data with mathematical modeling approaches to improve our understanding of NHP and human response to vaccine. We use a mathematical model to describe T-cell priming, proliferation, and differentiation in lymph nodes and blood, and calibrate the model to NHP and human blood data. Using the model, we demonstrate the impact of BCG timing on H56 vaccination response and reveal a general immunogenic response to H56 following BCG prime. Further, we use uncertainty and sensitivity analyses to isolate mechanisms driving differences in vaccination response observed between NHP and human datasets. This study highlights the power of a systems biology approach: integration of multiple modalities to better understand a complex biological system.
Collapse
Affiliation(s)
- Louis R. Joslyn
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elsje Pienaar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Robert M. DiFazio
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Sara Suliman
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Benjamin M. Kagina
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Jennifer J. Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
30
|
Joslyn LR, Pienaar E, DiFazio RM, Suliman S, Kagina BM, Flynn JL, Scriba TJ, Linderman JJ, Kirschner DE. Integrating Non-human Primate, Human, and Mathematical Studies to Determine the Influence of BCG Timing on H56 Vaccine Outcomes. Front Microbiol 2018; 9:1898. [PMID: 30177934 PMCID: PMC6110197 DOI: 10.3389/fimmu.2018.01898] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/31/2018] [Indexed: 12/21/2022] Open
Abstract
Background Acute lung injury (ALI) is characterized by suppressed fibrinolytic activity in bronchoalveolar lavage fluid (BALF) attributed to elevated plasminogen activator inhibitor-1 (PAI-1). Restoring pulmonary fibrinolysis by delivering tissue-type plasminogen activator (tPA), urokinase plasminogen activator (uPA), and plasmin could be a promising approach. Objectives To systematically analyze the overall benefit of fibrinolytic therapy for ALI reported in preclinical studies. Methods We searched PubMed, Embase, Web of Science, and CNKI Chinese databases, and analyzed data retrieved from 22 studies for the beneficial effects of fibrinolytics on animal models of ALI. Results Both large and small animals were used with five routes for delivering tPA, uPA, and plasmin. Fibrinolytics significantly increased the fibrinolytic activity both in the plasma and BALF. Fibrin degradation products in BALF had a net increase of 408.41 ng/ml vs controls (P < 0.00001). In addition, plasma thrombin–antithrombin complexes increased 1.59 ng/ml over controls (P = 0.0001). In sharp contrast, PAI-1 level in BALF decreased 21.44 ng/ml compared with controls (P < 0.00001). Arterial oxygen tension was improved by a net increase of 15.16 mmHg, while carbon dioxide pressure was significantly reduced (11.66 mmHg, P = 0.0001 vs controls). Additionally, fibrinolytics improved lung function and alleviated inflammation response: the lung wet/dry ratio was decreased 1.49 (P < 0.0001 vs controls), lung injury score was reduced 1.83 (P < 0.00001 vs controls), and BALF neutrophils were lesser (3 × 104/ml, P < 0.00001 vs controls). The mortality decreased significantly within defined study periods (6 h to 30 days for mortality), as the risk ratio of death was 0.2-fold of controls (P = 0.0008). Conclusion We conclude that fibrinolytic therapy may be effective pharmaceutic strategy for ALI in animal models.
Collapse
Affiliation(s)
- Louis R Joslyn
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States.,Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elsje Pienaar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States.,Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Robert M DiFazio
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Sara Suliman
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Benjamin M Kagina
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
31
|
de Paula Oliveira Santos B, Trentini MM, Machado RB, Rúbia Nunes Celes M, Kipnis A, Petrovsky N, Junqueira-Kipnis AP. Advax4 delta inulin combination adjuvant together with ECMX, a fusion construct of four protective mTB antigens, induces a potent Th1 immune response and protects mice against Mycobacterium tuberculosis infection. Hum Vaccin Immunother 2018; 13:2967-2976. [PMID: 28937879 DOI: 10.1080/21645515.2017.1368598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Tuberculosis (TB) remains a main public health concern and 10.4 million new cases occurred in 2015 around the world. BCG is the only approved vaccine against TB, but has variable efficacy and new vaccines are needed. We developed two new mTB vaccine candidates based on the recombinant fusion proteins, rCMX and rECMX formulated with Advax4, a new combination adjuvant combining delta inulin, CpG oligonucleotide and murabutide. BALB/c mice were immunized three times intramuscularly with these vaccine formulations. Injection of Advax4 alone increased the percentage of lymphatic endothelial cells and activated macrophages (F480/CD11b+) in the draining lymph nodes consistent with a chemotactic adjuvant effect. Advax4+CMX and Advax4+ECMX induced the highest levels of IgG1 and IgG2a antibodies against rCMX and rECMX, respectively. Immunized mice challenged with Mycobacterium tuberculosis (Mtb) had increased vaccine-specific Th1 responses in the lungs together with reduced Mtb - associated alveolar damage, although only the Advax4+ECMX vaccine demonstrated significant reduction of lung bacterial load. This study confirmed Advax4+ECMX as a potential TB vaccine candidate, with potential for further optimization and clinical development.
Collapse
Affiliation(s)
- Bruno de Paula Oliveira Santos
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Monalisa Martins Trentini
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Renato Beilner Machado
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Mara Rúbia Nunes Celes
- b Laboratory of Pathology, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - André Kipnis
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Nikolai Petrovsky
- c Flinders University and Vaxine Pty Ltd, Flinders Medical Center , Adelaide , Australia
| | - Ana Paula Junqueira-Kipnis
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| |
Collapse
|
32
|
Santoro F, Pettini E, Kazmin D, Ciabattini A, Fiorino F, Gilfillan GD, Evenroed IM, Andersen P, Pozzi G, Medaglini D. Transcriptomics of the Vaccine Immune Response: Priming With Adjuvant Modulates Recall Innate Responses After Boosting. Front Immunol 2018; 9:1248. [PMID: 29922291 PMCID: PMC5996052 DOI: 10.3389/fimmu.2018.01248] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Transcriptomic profiling of the immune response induced by vaccine adjuvants is of critical importance for the rational design of vaccination strategies. In this study, transcriptomics was employed to profile the effect of the vaccine adjuvant used for priming on the immune response following re-exposure to the vaccine antigen alone. Mice were primed with the chimeric vaccine antigen H56 of Mycobacterium tuberculosis administered alone or with the CAF01 adjuvant and boosted with the antigen alone. mRNA sequencing was performed on blood samples collected 1, 2, and 7 days after priming and after boosting. Gene expression analysis at day 2 after priming showed that the CAF01 adjuvanted vaccine induced a stronger upregulation of the innate immunity modules compared with the unadjuvanted formulation. The immunostimulant effect of the CAF01 adjuvant, used in the primary immunization, was clearly seen after a booster immunization with a low dose of antigen alone. One day after boost, we observed a strong upregulation of multiple genes in blood of mice primed with H56 + CAF01 compared with mice primed with the H56 alone. In particular, blood transcription modules related to innate immune response, such as monocyte and neutrophil recruitment, activation of antigen-presenting cells, and interferon response were activated. Seven days after boost, differential expression of innate response genes faded while a moderate differential expression of T cell activation modules was appreciable. Indeed, immunological analysis showed a higher frequency of H56-specific CD4+ T cells and germinal center B cells in draining lymph nodes, a strong H56-specific humoral response and a higher frequency of antibody-secreting cells in spleen of mice primed with H56 + CAF01. Taken together, these data indicate that the adjuvant used for priming strongly reprograms the immune response that, upon boosting, results in a stronger recall innate response essential for shaping the downstream adaptive response.
Collapse
Affiliation(s)
- Francesco Santoro
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Dmitri Kazmin
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Gregor D Gilfillan
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Ida M Evenroed
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Gianni Pozzi
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| |
Collapse
|
33
|
Gong W, Liang Y, Wu X. The current status, challenges, and future developments of new tuberculosis vaccines. Hum Vaccin Immunother 2018; 14:1697-1716. [PMID: 29601253 DOI: 10.1080/21645515.2018.1458806] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mycobacterium tuberculosis complex causes tuberculosis (TB), one of the top 10 causes of death worldwide. TB results in more fatalities than multi-drug resistant (MDR) HIV strain related coinfection. Vaccines play a key role in the prevention and control of infectious diseases. Unfortunately, the only licensed preventive vaccine against TB, bacilli Calmette-Guérin (BCG), is ineffective for prevention of pulmonary TB in adults. Therefore, it is very important to develop novel vaccines for TB prevention and control. This literature review provides an overview of the innate and adaptive immune response during M. tuberculosis infection, and presents current developments and challenges to novel TB vaccines. A comprehensive understanding of vaccines in preclinical and clinical studies provides extensive insight for the development of safer and more efficient vaccines, and may inspire new ideas for TB prevention and treatment.
Collapse
Affiliation(s)
- Wenping Gong
- a Army Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research , Haidian District, Beijing , China
| | - Yan Liang
- a Army Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research , Haidian District, Beijing , China
| | - Xueqiong Wu
- a Army Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research , Haidian District, Beijing , China
| |
Collapse
|
34
|
Platteel ACM, Nieuwenhuizen NE, Domaszewska T, Schürer S, Zedler U, Brinkmann V, Sijts AJAM, Kaufmann SHE. Efficacy Testing of H56 cDNA Tattoo Immunization against Tuberculosis in a Mouse Model. Front Immunol 2017; 8:1744. [PMID: 29312295 PMCID: PMC5732355 DOI: 10.3389/fimmu.2017.01744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/23/2017] [Indexed: 11/29/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a global threat. The only approved vaccine against TB, Mycobacterium bovis bacillus Calmette–Guérin (BCG), provides insufficient protection and, being a live vaccine, can cause disseminated disease in immunocompromised individuals. Previously, we found that intradermal cDNA tattoo immunization with cDNA of tetanus toxoid fragment C domain 1 fused to cDNA of the fusion protein H56, comprising the Mtb antigens Ag85B, ESAT-6, and Rv2660c, induced antigen-specific CD8+ T cell responses in vivo. As cDNA tattoo immunization would be safer than a live vaccine in immunocompromised patients, we tested the protective efficacy of intradermal tattoo immunization against TB with H56 cDNA, as well as with H56_E, a construct optimized for epitope processing in a mouse model. As Mtb antigens can be used in combination with BCG to boost immune responses, we also tested the protective efficacy of heterologous prime-boost, using dermal tattoo immunization with H56_E cDNA to boost BCG immunization in mice. Dermal H56 and H56_E cDNA immunization induced H56-specific CD4+ and CD8+ T cell responses and Ag85B-specific IgG antibodies, but did not reduce bacterial loads, although immunization with H56_E ameliorated lung pathology. Both subcutaneous and intradermal immunization with BCG resulted in broad cellular immune responses, with increased frequencies of CD4+ T effector memory cells, T follicular helper cells, and germinal center B cells, and resulted in reduced bacterial loads and lung pathology. Heterologous vaccination with BCG/H56_E cDNA induced increased H56-specific CD4+ and CD8+ T cell cytokine responses compared to vaccination with BCG alone, and lung pathology was significantly decreased in BCG/H56_E cDNA immunized mice compared to unvaccinated controls. However, bacterial loads were not decreased after heterologous vaccination compared to BCG alone. CD4+ T cells responding to Ag85B- and ESAT-6-derived epitopes were predominantly IFN-γ+TNF-α+ and TNF-α+IL-2+, respectively. In conclusion, despite inducing appreciable immune responses to Ag85B and ESAT-6, intradermal H56 cDNA tattoo immunization did not substantially enhance the protective effect of BCG under the conditions tested.
Collapse
Affiliation(s)
- Anouk C M Platteel
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | - Teresa Domaszewska
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Stefanie Schürer
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Ulrike Zedler
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Volker Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Alice J A M Sijts
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
35
|
Speth MT, Repnik U, Müller E, Spanier J, Kalinke U, Corthay A, Griffiths G. Poly(I:C)-Encapsulating Nanoparticles Enhance Innate Immune Responses to the Tuberculosis Vaccine Bacille Calmette-Guérin (BCG) via Synergistic Activation of Innate Immune Receptors. Mol Pharm 2017; 14:4098-4112. [PMID: 28974092 DOI: 10.1021/acs.molpharmaceut.7b00795] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The attenuated live vaccine strain bacille Calmette-Guérin (BCG) is currently the only available vaccine against tuberculosis (TB), but is largely ineffective against adult pulmonary TB, the most common disease form. This is in part due to BCG's ability to interfere with the host innate immune response, a feature that might be targeted to enhance the potency of this vaccine. Here, we investigated the ability of chitosan-based nanoparticles (pIC-NPs) containing polyinosinic-polycytidylic acid (poly(I:C)), an inducer of innate immunity via Toll-like receptor 3 (TLR3), to enhance the immunogenicity of BCG in mouse bone marrow derived macrophages (BMDM) in vitro. Incorporation of poly(I:C) into NPs protected it against degradation by ribonucleases and increased its uptake by mouse BMDM. Whereas soluble poly(I:C) was ineffective, pIC-NPs strongly enhanced the proinflammatory immune response of BCG-infected macrophages in a synergistic fashion, as evident by increased production of cytokines and induction of nitric oxide synthesis. Using macrophages from mice deficient in key signaling molecules involved in the pathogen recognition response, we identified combined activation of MyD88- and TRIF-dependent TLR signaling pathways to be essential for the synergistic effect between BCG and NP. Moreover, synergy was strongly dependent on the order of the two stimuli, with TLR activation by BCG functioning as the priming event for the subsequent pIC-NP stimulus, which acted through an auto-/paracrine type I interferon (IFN) feedback loop. Our results provide a foundation for a promising new approach to enhance BCG-vaccine immunogenicity by costimulation with NPs. They also contribute to a molecular understanding of the observed synergistic interaction between the pIC-NPs and BCG vaccine.
Collapse
Affiliation(s)
- Martin T Speth
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway
| | - Urska Repnik
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway
| | - Elisabeth Müller
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway.,Tumor Immunology lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo , N-0424 Oslo, Norway
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School , D-30625 Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School , D-30625 Hannover, Germany
| | - Alexandre Corthay
- Tumor Immunology lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo , N-0424 Oslo, Norway
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway
| |
Collapse
|
36
|
Abstract
INTRODUCTION Tuberculosis (TB) is an infectious disease caused mainly by Mycobacterium tuberculosis. In 2016, the WHO estimated 10.5 million new cases and 1.8 million deaths, making this disease the leading cause of death by an infectious agent. The current and projected TB situation necessitates the development of new vaccines with improved attributes compared to the traditional BCG method. Areas covered: In this review, the authors describe the most promising candidate vaccines against TB and discuss additional key elements in vaccine development, such as animal models, new adjuvants and immunization routes and new strategies for the identification of candidate vaccines. Expert opinion: At present, around 13 candidate vaccines for TB are in the clinical phase of evaluation; however, there is still no substitute for the BCG vaccine. One major impediment to developing an effective vaccine is our lack of understanding of several of the mechanisms associated with infection and the immune response against TB. However, the recent implementation of an entirely new set of technological advances will facilitate the proposal of new candidates. Finally, development of a new vaccine will require a major coordination of effort in order to achieve its effective administration to the people most in need of it.
Collapse
|
37
|
Mancha-Agresti P, de Castro CP, Dos Santos JSC, Araujo MA, Pereira VB, LeBlanc JG, Leclercq SY, Azevedo V. Recombinant Invasive Lactococcus lactis Carrying a DNA Vaccine Coding the Ag85A Antigen Increases INF-γ, IL-6, and TNF-α Cytokines after Intranasal Immunization. Front Microbiol 2017; 8:1263. [PMID: 28744263 PMCID: PMC5504179 DOI: 10.3389/fmicb.2017.01263] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/23/2017] [Indexed: 11/29/2022] Open
Abstract
Tuberculosis (TB) remains a major threat throughout the world and in 2015 it caused the death of 1.4 million people. The Bacillus Calmette-Guérin is the only existing vaccine against this ancient disease; however, it does not provide complete protection in adults. New vaccines against TB are eminently a global priority. The use of bacteria as vehicles for delivery of vaccine plasmids is a promising vaccination strategy. In this study, we evaluated the use of, an engineered invasive Lactococcus lactis (expressing Fibronectin-Binding Protein A from Staphylococcus aureus) for the delivery of DNA plasmid to host cells, especially to the mucosal site as a new DNA vaccine against tuberculosis. One of the major antigens documented that offers protective responses against Mycobacterium tuberculosis is the Ag85A. L. lactis FnBPA+ (pValac:Ag85A) which was obtained and used for intranasal immunization of C57BL/6 mice and the immune response profile was evaluated. In this study we observed that this strain was able to produce significant increases in the amount of pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-6) in the stimulated spleen cell supernatants, showing a systemic T helper 1 (Th1) cell response. Antibody production (IgG and sIgA anti-Ag85A) was also significantly increased in bronchoalveolar lavage, as well as in the serum of mice. In summary, these findings open new perspectives in the area of mucosal DNA vaccine, against specific pathogens using a Lactic Acid Bacteria such as L. lactis.
Collapse
Affiliation(s)
- Pamela Mancha-Agresti
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Camila Prosperi de Castro
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Janete S C Dos Santos
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel DiasBelo Horizonte, Brazil
| | - Maíra A Araujo
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel DiasBelo Horizonte, Brazil
| | - Vanessa B Pereira
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Jean G LeBlanc
- Centro de Referencia para Lactobacilos - Consejo Nacional de Investigaciones Científicas y TécnicasSan Miguel de Tucumán, Argentina
| | - Sophie Y Leclercq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel DiasBelo Horizonte, Brazil
| | - Vasco Azevedo
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| |
Collapse
|
38
|
Yu Q, Wang X, Fan X. A New Adjuvant MTOM Mediates Mycobacterium tuberculosis Subunit Vaccine to Enhance Th1-Type T Cell Immune Responses and IL-2 + T Cells. Front Immunol 2017; 8:585. [PMID: 28572807 PMCID: PMC5436497 DOI: 10.3389/fimmu.2017.00585] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/02/2017] [Indexed: 02/04/2023] Open
Abstract
The only licensed vaccine Mycobacterium bovis Bacillus Calmette–Guérin (BCG) cannot prevent the prevalence of tuberculosis (TB), which remains a major public health problem worldwide. A more effective TB vaccine than BCG is urgently needed. Subunit vaccine is a promising strategy, and suitable adjuvants will benefit the development of effective TB subunit vaccines. MTO, consisting of monophosphoryl lipid A (MPLA), trehalose-6,6′-dibehenate (TDB), and MF59, was developed as an adjuvant of TB vaccine because of its ability to evoke the Th1-type T cell responses, while it is insufficient to induce single and multifunctional IL-2+ T cells and has a limited ability to confer protection against Mycobacterium tuberculosis infection. Heat-killed Mycobacterium vaccae (Mv), which can evoke cytotoxic CD8+ and CD4+ T cell responses and has adjuvanticity, was, in this study, combined with MTO to produce a new adjuvant, called MTOM. The TB fusion protein Rv3407-PhoY2-Ag85A-Rv2626c-RpfB (WH121) was mixed with MTO, Mv, and MTOM to produce three subunit vaccines, and the protective efficacy and immune responses were compared in C57BL/6 mice. WH121/MTOM provided better protection against TB than the other two vaccines, matching the performance of BCG vaccine. MTOM showed stronger ability to increase single and multifunctional IL-2+ T cells and induce Th1-type responses than MTO or Mv. Therefore, MTOM might be a promising adjuvant that could contribute to the development of TB subunit vaccines.
Collapse
Affiliation(s)
- Qi Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.,School of Basic Medicine, Guiyang Traditional Chinese Medical College, Guiyang, China
| | - Xiaochun Wang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
39
|
Tkachuk AP, Gushchin VA, Potapov VD, Demidenko AV, Lunin VG, Gintsburg AL. Multi-subunit BCG booster vaccine GamTBvac: Assessment of immunogenicity and protective efficacy in murine and guinea pig TB models. PLoS One 2017; 12:e0176784. [PMID: 28453555 PMCID: PMC5409163 DOI: 10.1371/journal.pone.0176784] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/17/2017] [Indexed: 11/18/2022] Open
Abstract
New innovative vaccines are highly needed to combat the global threat posed by tuberculosis. Efficient components-antigens and adjuvants-are crucial for development of modern recombinant TB vaccines. This study describes a new vaccine (GamTBvac) consisting of two mycobacterial antigen fusions (Ag85A and ESAT6-CFP10)-with dextran-binding domain immobilized on dextran and mixed with an adjuvant consisting of DEAE-dextran core, and with CpG oligodeoxynucleotides (TLR9 agonists). GamTBvac and its components were assessed for immunogenicity and protective efficacy in GamTBvac-prime/boost and BCG-prime/ GamTBvac-boost in murine and guinea pig TB models. Results show that in both infectious models, GamTBvac has a strong immunogenicity and significant protective effect against Mycobacterium tuberculosis strain H37Rv under aerosol and intravenous challenges. GamTBvac showed a particularly strong protective effect as a BCG booster vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intravenous
- Aerosols
- Animals
- Antibodies, Bacterial/blood
- BCG Vaccine/immunology
- Cell Proliferation/physiology
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Female
- Guinea Pigs
- Immunization
- Immunization, Secondary
- Immunogenicity, Vaccine
- Lung/immunology
- Lymph Nodes/immunology
- Male
- Mice, Inbred C57BL
- Mycobacterium tuberculosis/immunology
- Spleen/immunology
- T-Lymphocytes/immunology
- Tuberculosis/immunology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- A. P. Tkachuk
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. A. Gushchin
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - V. D. Potapov
- State Research Center for Applied Microbiology and Biotechnology, Obolensk, Russia
| | - A. V. Demidenko
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. G. Lunin
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - A. L. Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
40
|
Moliva JI, Turner J, Torrelles JB. Immune Responses to Bacillus Calmette-Guérin Vaccination: Why Do They Fail to Protect against Mycobacterium tuberculosis? Front Immunol 2017; 8:407. [PMID: 28424703 PMCID: PMC5380737 DOI: 10.3389/fimmu.2017.00407] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis (TB), is the current leading cause of death due to a single infectious organism. Although curable, the broad emergence of multi-, extensive-, extreme-, and total-drug resistant strains of M.tb has hindered eradication efforts of this pathogen. Furthermore, computational models predict a quarter of the world’s population is infected with M.tb in a latent state, effectively serving as the largest reservoir for any human pathogen with the ability to cause significant morbidity and mortality. The World Health Organization has prioritized new strategies for improved vaccination programs; however, the lack of understanding of mycobacterial immunity has made it difficult to develop new successful vaccines. Currently, Mycobacterium bovis bacillus Calmette–Guérin (BCG) is the only vaccine approved for use to prevent TB. BCG is highly efficacious at preventing meningeal and miliary TB, but is at best 60% effective against the development of pulmonary TB in adults and wanes as we age. In this review, we provide a detailed summary on the innate immune response of macrophages, dendritic cells, and neutrophils in response to BCG vaccination. Additionally, we discuss adaptive immune responses generated by BCG vaccination, emphasizing their specific contributions to mycobacterial immunity. The success of future vaccines against TB will directly depend on our understanding of mycobacterial immunity.
Collapse
Affiliation(s)
- Juan I Moliva
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
41
|
Wang J, Liu R, Liu B, Yang Y, Xie J, Zhu N. Systems Pharmacology-based strategy to screen new adjuvant for hepatitis B vaccine from Traditional Chinese Medicine Ophiocordyceps sinensis. Sci Rep 2017; 7:44788. [PMID: 28317886 PMCID: PMC5357901 DOI: 10.1038/srep44788] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 02/14/2017] [Indexed: 12/19/2022] Open
Abstract
Adjuvants are common component for many vaccines but there are still few licensed for human use due to low efficiency or side effects. The present work adopted Systems Pharmacology analysis as a new strategy to screen adjuvants from traditional Chinese medicine. Ophiocordyceps sinensis has been used for many years in China and other Asian countries with many biological properties, but the pharmacological mechanism has not been fully elucidated. First in this study, 190 putative targets for 17 active compounds in Ophiocordyceps sinensis were retrieved and a systems pharmacology-based approach was applied to provide new insights into the pharmacological actions of the drug. Pathway enrichment analysis found that the targets participated in several immunological processes. Based on this, we selected cordycepin as a target compound to serve as an adjuvant of the hepatitis B vaccine because the existing vaccine often fails to induce an effective immune response in many subjects. Animal and cellular experiments finally validated that the new vaccine simultaneously improves the humoral and cellular immunity of BALB/c mice without side effects. All this results demonstrate that cordycepin could work as adjuvant to hepatitis b vaccine and systems-pharmacology analysis could be used as a new method to select adjuvants.
Collapse
Affiliation(s)
- Jingbo Wang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, Institute of Biomedical Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Rui Liu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, Institute of Biomedical Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Baoxiu Liu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, Institute of Biomedical Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yan Yang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, Institute of Biomedical Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jun Xie
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, Institute of Biomedical Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Naishuo Zhu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, Institute of Biomedical Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| |
Collapse
|
42
|
Marques Neto LM, Kipnis A, Junqueira-Kipnis AP. Role of Metallic Nanoparticles in Vaccinology: Implications for Infectious Disease Vaccine Development. Front Immunol 2017; 8:239. [PMID: 28337198 PMCID: PMC5340775 DOI: 10.3389/fimmu.2017.00239] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/20/2017] [Indexed: 02/04/2023] Open
Abstract
Subunit vaccines are safer but less immunogenic than live-attenuated vaccines or whole cell inactivated vaccines. Adjuvants are used to enhance and modulate antigen (Ag) immunogenicity, aiming to induce a protective and long-lasting immune response. Several molecules and formulations have been studied for their adjuvanticity, but only seven have been approved to formulate human vaccines. Metallic nanoparticles (MeNPs), particularly those containing gold and iron oxides, are widely used in medicine for diagnosis and therapy and have been used as carriers for drugs and vaccines. However, little is known about the immune response elicited by MeNPs or about their importance in the development of new vaccines. There is evidence that these particles display adjuvant characteristics, promoting cell recruitment, antigen-presenting cell activation, cytokine production, and inducing a humoral immune response. This review focuses on the characteristics of MeNPs that could facilitate the induction of a cellular immune response, particularly T-helper 1 and T-helper 17, and their potential functions as adjuvants for subunit vaccines.
Collapse
Affiliation(s)
- Lázaro Moreira Marques Neto
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia, Goiás , Brazil
| | - André Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia, Goiás , Brazil
| | - Ana Paula Junqueira-Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia, Goiás , Brazil
| |
Collapse
|
43
|
Omp16-based vaccine encapsulated by alginate-chitosan microspheres provides significant protection against Haemophilus parasuis in mice. Vaccine 2017; 35:1417-1423. [PMID: 28187951 DOI: 10.1016/j.vaccine.2017.01.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/23/2017] [Accepted: 01/26/2017] [Indexed: 12/13/2022]
Abstract
Haemophilus parasuis (H. parasuis) is the etiological agent of swine Glässer's disease, which leads to significant economic loss in swine industry over the world. Subunit vaccine based on outer membrane protein is one of the promising choices to protect pigs against H. parasuis infection despite low immunity efficiency. In this paper, outer membrane protein 16 (Omp16) of H. parasuis encapsulated by alginate-chitosan microspheres as antigen carriers was explored for the first time in a mouse model. Our results showed that the microspheres with Omp16 induced significant higher H. parasuis-specific antibodies, and higher titers of IL-2, IL-4, and IFN-γ than those by Omp16-FIA in treated mice (p<0.05). Moreover, H. parasuis load in the tissues from liver, spleen, and lung of mice immunized with microspheres containing Omp16 was significantly decreased (p<0.05) than that in the same counterpart tissues of control groups. In addition, 80% mice treated with Omp16 and 70% mice with Omp16-FIA were survived after challenged with H. parasuis virulent strain LY02 (serovar 5). Therefore, Omp16-based microsphere vaccine induces both humoral and cellular immune responses and provides promising protection against H. parasuis infection in mice.
Collapse
|
44
|
Ding Y, Zheng H, Feng C, Wang B, Liu C, Mi K, Cao H, Meng S. Heat-Shock Protein gp96 Enhances T Cell Responses and Protective Potential to Bacillus Calmette-Guérin Vaccine. Scand J Immunol 2017; 84:222-8. [PMID: 27417661 DOI: 10.1111/sji.12463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/12/2016] [Indexed: 01/28/2023]
Abstract
The commonly used Bacillus Calmette-Guérin (BCG) vaccine only induces moderate T cell responses and is less effective in protecting against pulmonary tuberculosis (TB) in adults and ageing populations. Thus, developing new TB vaccine candidates is an important strategy against the spread of Mycobacterium tuberculosis. Here, we demonstrated that immunization with heat-shock protein gp96 as an adjuvant led to a significantly increased CD4(+) and CD8(+) T cell response to a BCG vaccine. Secretion of the Th1-type cytokines was increased by splenocytes from gp96-immunized mice. In addition, adding gp96 as an adjuvant effectively improved the protection against intravenous challenge with Mycobacterium bovis BCG in mice. Our study reveals the novel property of gp96 in boosting the vaccine-specific T cell response and its potential use as an adjuvant for BCG vaccines against mycobacterial infection.
Collapse
Affiliation(s)
- Y Ding
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - H Zheng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - C Feng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - B Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - C Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - K Mi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - H Cao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China.
| | - S Meng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
| |
Collapse
|
45
|
Immunogenic Properties of Lactobacillus plantarum Producing Surface-Displayed Mycobacterium tuberculosis Antigens. Appl Environ Microbiol 2016; 83:AEM.02782-16. [PMID: 27815271 DOI: 10.1128/aem.02782-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/27/2016] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis (TB) remains among the most deadly diseases in the world. The only available vaccine against tuberculosis is the bacille Calmette-Guérin (BCG) vaccine, which does not ensure full protection in adults. There is a global urgency for the development of an effective vaccine for preventing disease transmission, and it requires novel approaches. We are exploring the use of lactic acid bacteria (LAB) as a vector for antigen delivery to mucosal sites. Here, we demonstrate the successful expression and surface display of a Mycobacterium tuberculosis fusion antigen (comprising Ag85B and ESAT-6, referred to as AgE6) on Lactobacillus plantarum The AgE6 fusion antigen was targeted to the bacterial surface using two different anchors, a lipoprotein anchor directing the protein to the cell membrane and a covalent cell wall anchor. AgE6-producing L. plantarum strains using each of the two anchors induced antigen-specific proliferative responses in lymphocytes purified from TB-positive donors. Similarly, both strains induced immune responses in mice after nasal or oral immunization. The impact of the anchoring strategies was reflected in dissimilarities in the immune responses generated by the two L. plantarum strains in vivo The present study comprises an initial step toward the development of L. plantarum as a vector for M. tuberculosis antigen delivery. IMPORTANCE This work presents the development of Lactobacillus plantarum as a candidate mucosal vaccine against tuberculosis. Tuberculosis remains one of the top infectious diseases worldwide, and the only available vaccine, bacille Calmette-Guérin (BCG), fails to protect adults and adolescents. Direct antigen delivery to mucosal sites is a promising strategy in tuberculosis vaccine development, and lactic acid bacteria potentially provide easy, safe, and low-cost delivery vehicles for mucosal immunization. We have engineered L. plantarum strains to produce a Mycobacterium tuberculosis fusion antigen and to anchor this antigen to the bacterial cell wall or to the cell membrane. The recombinant strains elicited proliferative antigen-specific T-cell responses in white blood cells from tuberculosis-positive humans and induced specific immune responses after nasal and oral administrations in mice.
Collapse
|
46
|
Triccas JA, Counoupas C. Novel vaccination approaches to prevent tuberculosis in children. Pneumonia (Nathan) 2016; 8:18. [PMID: 28702297 PMCID: PMC5471729 DOI: 10.1186/s41479-016-0020-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
Pediatric tuberculosis (TB) is an underappreciated problem and accounts for 10 % of all TB deaths worldwide. Children are highly susceptible to infection with Mycobacterium tuberculosis and interrupting TB spread would require the development of effective strategies to control TB transmission in pediatric populations. The current vaccine for TB, M. bovis Bacille Calmette-Guérin (BCG), can afford some level of protection against TB meningitis and severe forms of disseminated TB in children; however, its efficacy against pulmonary TB is variable and the vaccine does not afford life-long protective immunity. For these reasons there is considerable interest in the development of new vaccines to control TB in children. Multiple vaccine strategies are being assessed and include recombinant forms of the existing BCG vaccine, protein or viral candidates designed to boost BCG-induced immunity, or live attenuated forms of M. tuberculosis. A number of these candidates have entered clinical trials; however, no vaccine has shown improved protective efficacy compared to BCG in humans. The current challenge is to identify the most suitable candidates to progress from early to late stage clinical trials, in order to deliver a vaccine that can control and hopefully eliminate the global threat of TB.
Collapse
Affiliation(s)
- James A Triccas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Level 5, Charles Perkins Centre D17, Sydney, NSW 2006 Australia.,Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW Australia.,Sydney Medical School and the Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW Australia
| | - Claudio Counoupas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Level 5, Charles Perkins Centre D17, Sydney, NSW 2006 Australia.,Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW Australia
| |
Collapse
|