101
|
Baldwin SL, Reese VA, Larsen SE, Beebe E, Guderian J, Orr MT, Fox CB, Reed SG, Coler RN. Prophylactic efficacy against Mycobacterium tuberculosis using ID93 and lipid-based adjuvant formulations in the mouse model. PLoS One 2021; 16:e0247990. [PMID: 33705411 PMCID: PMC7951850 DOI: 10.1371/journal.pone.0247990] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/17/2021] [Indexed: 11/19/2022] Open
Abstract
An estimated 10 million people developed tuberculosis (TB) disease in 2019 which underscores the need for a vaccine that prevents disease and reduces transmission. The aim of our current studies is to characterize and test a prophylactic tuberculosis vaccine comprised of ID93, a polyprotein fusion antigen, and a liposomal formulation [including a synthetic TLR4 agonist (glucopyranosyl lipid adjuvant, GLA) and QS-21] in a preclinical mouse model of TB disease. Comparisons of the ID93+GLA-LSQ vaccines are also made to the highly characterized ID93+GLA-SE oil-in-water emulsion adjuvant, which are also included these studies. The recent success of vaccine candidate M72 combined with adjuvant AS01E (GlaxoSmithKline Biologicals) in reducing progression to active disease is promising and has renewed excitement for experimental vaccines currently in the TB vaccine pipeline. The AS01E adjuvant contains monophosphoryl lipid A (MPL) and QS-21 (a saponin) in a liposomal formulation. While AS01E has demonstrated potent adjuvant activity as a component of both approved and experimental vaccines, developing alternatives to this adjuvant system will become important to fill the high demand envisioned for future vaccine needs. Furthermore, replacement sources of potent adjuvants will help to supply the demand of a TB vaccine [almost one-quarter of the world's population are estimated to have latent Mycobacterium tuberculosis (Mtb) according to the WHO 2019 global TB report], addressing (a) cost of goods, (b) supply of goods, and (c) improved efficacy of subunit vaccines against Mtb. We show that both ID93+GLA-SE (containing an emulsion adjuvant) and ID93+GLA-LSQ (containing a liposomal adjuvant) induce ID93-specific TH1 cellular immunity including CD4+CD44+ T cells expressing IFNγ, TNF, and IL-2 (using flow cytometry and intracellular cytokine staining) and vaccine-specific IgG2 antibody responses (using an ELISA). In addition, both ID93+GLA-SE and ID93+GLA-LSQ effectively decrease the bacterial load within the lungs of mice infected with Mtb. Formulations based on this liposomal adjuvant formulation may provide an alternative to AS01 adjuvant systems.
Collapse
Affiliation(s)
- Susan L. Baldwin
- Seattle Children’s Research Institute, Seattle, WA, United States of America
- * E-mail:
| | - Valerie A. Reese
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sasha E. Larsen
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Elyse Beebe
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Jeff Guderian
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Mark T. Orr
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Christopher B. Fox
- Infectious Disease Research Institute, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Rhea N. Coler
- Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
102
|
Tiam ER, Bikobo DSN, Ndassa IM, Nyemeck Ii NM, A Zintchem AA, Ayong L, Diboué PHB, Ndjakou BL, Mbing JN, Pegnyemb DE. Experimental and computational studies of an antiplasmodial derivative of allantoin; antimycobacterial essential oil from Cordia batesii WERNHAM (Boraginaceae). BMC Chem 2021; 15:15. [PMID: 33673871 PMCID: PMC7934435 DOI: 10.1186/s13065-021-00742-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background Chemical and pharmacological investigations were performed on the stems of Cordia batesii (Boraginaeae); chemical studies included quantum calculations applied on a newly described compound. Results A new derivative of allantoin (1) named batesiin (2) was characterized. Thirteen other known compounds involving allantoin (1) were either isolated or identified. GC–MS enabled the identification of six compounds from a fraction containing essential oil. MeOH extract and some isolated compounds were tested in vitro against Pf7G8 CQS and Pf Dd2 CQR strains of Plasmodium falciparum; extract disclosed a moderate antiplasmodial activity (IC50 = 50 μg mL−1). Meantime, the CH2Cl2 extract and essential oil fraction were tested on a resistant mycobacterial strain of Mycobacterium tuberculosis; a potent antimycobacterial activity with a MIC = 9.52 μg mL−1 was deduced from essential oil. Density functional theory (DFT) calculations were carried on batesiin (2). Calculated chemical shifts at B3LYP/6-31G(d,p) and MPW1PW91/6-31G+(d,p) showed much better correlations with the experimental data. Time dependent DFT at B3LYP/6-31G+(d,p) displayed a major absorption band 3.01 nm higher than the experimental value. Conclusion Cordia batesii can be considered as promising in search of compounds with antimalarial and antitubercular properties. DFT studies are very helpful when trying to learn more about the spectroscopic insights of a derivative of allantoin (1).
Collapse
Affiliation(s)
- Eric Robert Tiam
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon
| | | | - Ibrahim Mbouombouo Ndassa
- Department of Inorganic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon.,Department of Chemistry, Higher Training College, University of Yaoundé I, P.O Box 47, Yaounde, Cameroon
| | - Norbert Mbabi Nyemeck Ii
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon
| | - Auguste Abouem A Zintchem
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon. .,Department of Chemistry, Higher Training College, University of Yaoundé I, P.O Box 47, Yaounde, Cameroon.
| | | | - Patrick Hervé Betote Diboué
- Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Bruno Lenta Ndjakou
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon.,Department of Chemistry, Higher Training College, University of Yaoundé I, P.O Box 47, Yaounde, Cameroon
| | - Joséphine Ngo Mbing
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon
| | - Dieudonné Emmanuel Pegnyemb
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O Box 812, Yaounde, Cameroon
| |
Collapse
|
103
|
Netikul T, Palittapongarnpim P, Thawornwattana Y, Plitphonganphim S. Estimation of the global burden of Mycobacterium tuberculosis lineage 1. INFECTION GENETICS AND EVOLUTION 2021; 91:104802. [PMID: 33684570 DOI: 10.1016/j.meegid.2021.104802] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/16/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022]
Abstract
Tuberculosis is still problematic as it affects large numbers of people globally. Mycobacterium tuberculosis Lineage 1 (L1) or Indo Oceanic Lineage, one of widespread major lineages, has a specific geographic distribution and high mortality. It is highly diverse and endemic in several high burden countries. However, studies on the global burden of L1 and its sublineages remain limited. This may lead to the underestimation of the importance of its variance in developing and applying tuberculosis control measures. This study aimed to estimate the number of patients infected with M. tuberculosis L1 and its sublineages worldwide. The proportion of L1 among tuberculosis patients was searched in published reports from countries around the world and the number of patients was calculated based on a WHO report on country incidences and populations. The numbers of patients infected with the five major sublineages, namely L1.1.1, L1.1.2, L1.1.3, L1.2.1, and L1.2.2 were estimated where information was available. It was found that L1 accounted for 28% of global tuberculosis cases in 2012 and 2018. Over 80% of the L1 global burden was in India, the Philippines, Indonesia and Bangladesh, which are also among the countries with highest absolute numbers of tuberculosis patients in the world. Globally, the estimated number of patients infected with M. tuberculosis L1.2.1 and L1.1.2 was over 1.1 million and of patients infected with L1.1.1 was about 200,000. This study demonstrated that L1 contributes significantly to the global burden of tuberculosis. To achieve the End TB Strategy, more attention needs to be paid to the responses of M. tuberculosis L1 to various control measures.
Collapse
Affiliation(s)
- Thidarat Netikul
- Faculty of Medicine, Siam University, Phet Kasem Road, Bangkok, Thailand
| | - Prasit Palittapongarnpim
- Pornchai Matangkasombut Center for Microbial Genomics, Department of Microbiology, Faculty of Science, Mahidol University, Rama 6 road, Bangkok, Thailand; National Science and Technology Development Agency, Pathumthani, Thailand
| | - Yuttapong Thawornwattana
- Pornchai Matangkasombut Center for Microbial Genomics, Department of Microbiology, Faculty of Science, Mahidol University, Rama 6 road, Bangkok, Thailand
| | - Supada Plitphonganphim
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Rama 6 road, Bangkok, Thailand.
| |
Collapse
|
104
|
Fatma F, Tripathi DK, Srivastava M, Srivastava KK, Arora A. Immunological characterization of chimeras of high specificity antigens from Mycobacterium tuberculosis H37Rv. Tuberculosis (Edinb) 2021; 127:102054. [PMID: 33550109 DOI: 10.1016/j.tube.2021.102054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
Abstract
Tuberculosis remains a serious global health problem. BCG is the only prophylactic TB vaccine and it shows variable protective efficacy. Chimeric protein subunit vaccines hold great potential as stand-alone vaccines or heterologous BCG prime boosters. We have designed a protein chimera, PP31, by combining Mtb ESAT-6 family antigen Rv1198 and MoCo biosynthesis family antigen Rv3111. Further, PP31 was extended by addition of latency antigen Rv1813c to yield PP43. Immunization of BALB/c mice with PP31 or PP43 with FIA adjuvant elicited strong humoral immune response. Restimulation of splenocytes of the immunized mice lead to significant proliferation of lymphocytes, secretion of cytokines IFN-γ, TNF, IL-2 of the Th1 class, IL-17A of the Th17 class, and IL-6. PP31 and PP43 also induced intracellular cytokine expression (IFN-γ, TNF, and IL-2) from both CD4+-CD44high and CD8+-CD44high T-cells. Antigen-specific IFN-γ+/IL-2+ double positive CD4+ T-cells were significantly higher in case of PP43 than PP31-immunized mice and control group. PP43 showed protection equivalent to heat-inactivated BCG in response to challenge of the immunized mice with Mtb H37Ra. Based on its immunogenicity and protective efficacy, PP43 appears to be a potential candidate for further development as a subunit vaccine against TB.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/immunology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytokines/metabolism
- Epitopes
- Female
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Immunogenicity, Vaccine
- Lymphocyte Activation/drug effects
- Lymphocyte Subsets/drug effects
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Mice, Inbred BALB C
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/immunology
- Tuberculosis/blood
- Tuberculosis/immunology
- Tuberculosis/microbiology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Mice
Collapse
Affiliation(s)
- Farheen Fatma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Dinesh K Tripathi
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Mrigank Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Kishore K Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
| | - Ashish Arora
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
105
|
Weerasuriya CK, Harris RC, McQuaid CF, Bozzani F, Ruan Y, Li R, Li T, Rade K, Rao R, Ginsberg AM, Gomez GB, White RG. The epidemiologic impact and cost-effectiveness of new tuberculosis vaccines on multidrug-resistant tuberculosis in India and China. BMC Med 2021; 19:60. [PMID: 33632218 PMCID: PMC7908776 DOI: 10.1186/s12916-021-01932-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Despite recent advances through the development pipeline, how novel tuberculosis (TB) vaccines might affect rifampicin-resistant and multidrug-resistant tuberculosis (RR/MDR-TB) is unknown. We investigated the epidemiologic impact, cost-effectiveness, and budget impact of hypothetical novel prophylactic prevention of disease TB vaccines on RR/MDR-TB in China and India. METHODS We constructed a deterministic, compartmental, age-, drug-resistance- and treatment history-stratified dynamic transmission model of tuberculosis. We introduced novel vaccines from 2027, with post- (PSI) or both pre- and post-infection (P&PI) efficacy, conferring 10 years of protection, with 50% efficacy. We measured vaccine cost-effectiveness over 2027-2050 as USD/DALY averted-against 1-times GDP/capita, and two healthcare opportunity cost-based (HCOC), thresholds. We carried out scenario analyses. RESULTS By 2050, the P&PI vaccine reduced RR/MDR-TB incidence rate by 71% (UI: 69-72) and 72% (UI: 70-74), and the PSI vaccine by 31% (UI: 30-32) and 44% (UI: 42-47) in China and India, respectively. In India, we found both USD 10 P&PI and PSI vaccines cost-effective at the 1-times GDP and upper HCOC thresholds and P&PI vaccines cost-effective at the lower HCOC threshold. In China, both vaccines were cost-effective at the 1-times GDP threshold. P&PI vaccine remained cost-effective at the lower HCOC threshold with 49% probability and PSI vaccines at the upper HCOC threshold with 21% probability. The P&PI vaccine was predicted to avert 0.9 million (UI: 0.8-1.1) and 1.1 million (UI: 0.9-1.4) second-line therapy regimens in China and India between 2027 and 2050, respectively. CONCLUSIONS Novel TB vaccination is likely to substantially reduce the future burden of RR/MDR-TB, while averting the need for second-line therapy. Vaccination may be cost-effective depending on vaccine characteristics and setting.
Collapse
Affiliation(s)
- Chathika K Weerasuriya
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| | - Rebecca C Harris
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, UK.,Currently employed at Sanofi Pasteur, Singapore, Singapore
| | - C Finn McQuaid
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Fiammetta Bozzani
- Department of Global Health and Development, Faculty of Public Health & Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Yunzhou Ruan
- Chinese Centre for Disease Control and Prevention, Beijing, China
| | - Renzhong Li
- Chinese Centre for Disease Control and Prevention, Beijing, China
| | - Tao Li
- Chinese Centre for Disease Control and Prevention, Beijing, China
| | | | - Raghuram Rao
- National Tuberculosis Elimination Programme, New Delhi, India
| | - Ann M Ginsberg
- International AIDS Vaccine Initiative, New York, USA.,Current Affiliation: Bill and Melinda Gates Foundation, Washington DC, USA
| | - Gabriela B Gomez
- Department of Global Health and Development, Faculty of Public Health & Policy, London School of Hygiene and Tropical Medicine, London, UK.,Currently employed at Sanofi Pasteur, Lyon, France
| | - Richard G White
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
106
|
Ravesloot-Chávez MM, Van Dis E, Stanley SA. The Innate Immune Response to Mycobacterium tuberculosis Infection. Annu Rev Immunol 2021; 39:611-637. [PMID: 33637017 DOI: 10.1146/annurev-immunol-093019-010426] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infection with Mycobacterium tuberculosis causes >1.5 million deaths worldwide annually. Innate immune cells are the first to encounter M. tuberculosis, and their response dictates the course of infection. Dendritic cells (DCs) activate the adaptive response and determine its characteristics. Macrophages are responsible both for exerting cell-intrinsic antimicrobial control and for initiating and maintaining inflammation. The inflammatory response to M. tuberculosis infection is a double-edged sword. While cytokines such as TNF-α and IL-1 are important for protection, either excessive or insufficient cytokine production results in progressive disease. Furthermore, neutrophils-cells normally associated with control of bacterial infection-are emerging as key drivers of a hyperinflammatory response that results in host mortality. The roles of other innate cells, including natural killer cells and innate-like T cells, remain enigmatic. Understanding the nuances of both cell-intrinsic control of infection and regulation of inflammation will be crucial for the successful development of host-targeted therapeutics and vaccines.
Collapse
Affiliation(s)
| | - Erik Van Dis
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA; ,
| | - Sarah A Stanley
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA; , .,Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California 94720, USA
| |
Collapse
|
107
|
Rijnink WF, Ottenhoff THM, Joosten SA. B-Cells and Antibodies as Contributors to Effector Immune Responses in Tuberculosis. Front Immunol 2021; 12:640168. [PMID: 33679802 PMCID: PMC7930078 DOI: 10.3389/fimmu.2021.640168] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/29/2021] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is still a major threat to mankind, urgently requiring improved vaccination and therapeutic strategies to reduce TB-disease burden. Most present vaccination strategies mainly aim to induce cell-mediated immunity (CMI), yet a series of independent studies has shown that B-cells and antibodies (Abs) may contribute significantly to reduce the mycobacterial burden. Although early studies using B-cell knock out animals did not support a major role for B-cells, more recent studies have provided new evidence that B-cells and Abs can contribute significantly to host defense against Mtb. B-cells and Abs exist in many different functional subsets, each equipped with unique functional properties. In this review, we will summarize current evidence on the contribution of B-cells and Abs to immunity toward Mtb, their potential utility as biomarkers, and their functional contribution to Mtb control.
Collapse
Affiliation(s)
- Willemijn F Rijnink
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
108
|
Synthetic protein conjugate vaccines provide protection against Mycobacterium tuberculosis in mice. Proc Natl Acad Sci U S A 2021; 118:2013730118. [PMID: 33468674 DOI: 10.1073/pnas.2013730118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The global incidence of tuberculosis remains unacceptably high, with new preventative strategies needed to reduce the burden of disease. We describe here a method for the generation of synthetic self-adjuvanted protein vaccines and demonstrate application in vaccination against Mycobacterium tuberculosis Two vaccine constructs were designed, consisting of full-length ESAT6 protein fused to the TLR2-targeting adjuvants Pam2Cys-SK4 or Pam3Cys-SK4 These were produced by chemical synthesis using a peptide ligation strategy. The synthetic self-adjuvanting vaccines generated powerful local CD4+ T cell responses against ESAT6 and provided significant protection in the lungs from virulent M. tuberculosis aerosol challenge when administered to the pulmonary mucosa of mice. The flexible synthetic platform we describe, which allows incorporation of adjuvants to multiantigenic vaccines, represents a general approach that can be applied to rapidly assess vaccination strategies in preclinical models for a range of diseases, including against novel pandemic pathogens such as SARS-CoV-2.
Collapse
|
109
|
Minias A, Żukowska L, Lechowicz E, Gąsior F, Knast A, Podlewska S, Zygała D, Dziadek J. Early Drug Development and Evaluation of Putative Antitubercular Compounds in the -Omics Era. Front Microbiol 2021; 11:618168. [PMID: 33603720 PMCID: PMC7884339 DOI: 10.3389/fmicb.2020.618168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by the bacterium Mycobacterium tuberculosis. According to the WHO, the disease is one of the top 10 causes of death of people worldwide. Mycobacterium tuberculosis is an intracellular pathogen with an unusually thick, waxy cell wall and a complex life cycle. These factors, combined with M. tuberculosis ability to enter prolonged periods of latency, make the bacterium very difficult to eradicate. The standard treatment of TB requires 6-20months, depending on the drug susceptibility of the infecting strain. The need to take cocktails of antibiotics to treat tuberculosis effectively and the emergence of drug-resistant strains prompts the need to search for new antitubercular compounds. This review provides a perspective on how modern -omic technologies facilitate the drug discovery process for tuberculosis treatment. We discuss how methods of DNA and RNA sequencing, proteomics, and genetic manipulation of organisms increase our understanding of mechanisms of action of antibiotics and allow the evaluation of drugs. We explore the utility of mathematical modeling and modern computational analysis for the drug discovery process. Finally, we summarize how -omic technologies contribute to our understanding of the emergence of drug resistance.
Collapse
Affiliation(s)
- Alina Minias
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Lidia Żukowska
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- BioMedChem Doctoral School of the University of Lodz and the Institutes of the Polish Academy of Sciences in Lodz, Lodz, Poland
| | - Ewelina Lechowicz
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Filip Gąsior
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- BioMedChem Doctoral School of the University of Lodz and the Institutes of the Polish Academy of Sciences in Lodz, Lodz, Poland
| | - Agnieszka Knast
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Lodz, Poland
| | - Sabina Podlewska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Krakow, Poland
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Daria Zygała
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
- Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Jarosław Dziadek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
110
|
Hakim JMC, Yang Z. Predicted Structural Variability of Mycobacterium tuberculosis PPE18 Protein With Immunological Implications Among Clinical Strains. Front Microbiol 2021; 11:595312. [PMID: 33488541 PMCID: PMC7819968 DOI: 10.3389/fmicb.2020.595312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/27/2020] [Indexed: 11/13/2022] Open
Abstract
Recent advancements in vaccinology have led to the development of the M72/AS01E subunit vaccine, of which the major component is the Mycobacterium tuberculosis (MTB) PPE18 protein. Previous studies have demonstrated the genetic variability of the gene encoding PPE18 protein and the resulting peptide changes in diverse clinical strains of MTB; however, none have modeled the structural changes resulting from these peptide changes and their immunological implications. In this study, we investigated the structural predictions of 29 variant PPE18 proteins previously reported. We found evidence that PPE18 is at least a two-domain protein, with a highly conserved first domain and a largely variable second domain that has different coevolutionary clusters. Further, we investigated putative epitope sites in the clinical variants of PPE18 using prediction software. We found a negative relationship between T-cell epitope number and residue variability, while B-cell epitope likelihood was positively correlated with residue variability. Moreover, we found far more residues in the second domain predicted to be B-cell epitopes compared with the first domain. These results suggest an important functional role of the first domain and a role in immune evasion for the second, which extends our knowledge base of the basic biology of the PPE18 protein and indicates the need for further study into non-traditional immunological responses to TB.
Collapse
Affiliation(s)
- Jill M C Hakim
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Zhenhua Yang
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
111
|
Verrall AJ, Schneider M, Alisjahbana B, Apriani L, van Laarhoven A, Koeken VACM, van Dorp S, Diadani E, Utama F, Hannaway RF, Indrati A, Netea MG, Sharples K, Hill PC, Ussher JE, van Crevel R. Early Clearance of Mycobacterium tuberculosis Is Associated With Increased Innate Immune Responses. J Infect Dis 2021; 221:1342-1350. [PMID: 30958547 DOI: 10.1093/infdis/jiz147] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND A proportion of tuberculosis (TB) case contacts do not become infected, even when heavily exposed. We studied the innate immune responses of TB case contacts to understand their role in protection against infection with Mycobacterium tuberculosis, termed "early clearance." METHODS Indonesian household contacts of TB cases were tested for interferon-γ release assay (IGRA) conversion between baseline and 14 weeks post recruitment. Blood cell populations and ex vivo innate whole blood cytokine responses were measured at baseline and, in a subgroup, flow cytometry was performed at weeks 2 and 14. Immunological characteristics were measured for early clearers, defined as a persistently negative IGRA at 3 months, and converters, whose IGRA converted from negative to positive. RESULTS Among 1347 case contacts, 317 were early clearers and 116 were converters. Flow cytometry showed a resolving innate cellular response from 2 to 14 weeks in persistently IGRA-negative contacts but not converters. There were no differences in cytokine responses to mycobacterial stimuli, but compared to converters, persistently IGRA-negative contacts produced more proinflammatory cytokines following heterologous stimulation with Escherichia coli and Streptococcus pneumoniae. CONCLUSIONS Early clearance of M. tuberculosis is associated with enhanced heterologous innate immune responses similar to those activated during induction of trained immunity.
Collapse
Affiliation(s)
- Ayesha J Verrall
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - Marion Schneider
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Bachti Alisjahbana
- TB-HIV Research Center, Faculty of Medicine, Universitas Padjadjaran, Nijmegen, The Netherlands.,Department of Internal Medicine, Faculty of Medicine, Universitas Padajdaran, Hasan Sadikin Hospital, Nijmegen, The Netherlands
| | - Lika Apriani
- TB-HIV Research Center, Faculty of Medicine, Universitas Padjadjaran, Nijmegen, The Netherlands.,Department of Public Health, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Arjan van Laarhoven
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, The Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, The Netherlands
| | - Suszanne van Dorp
- Department of Haematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emira Diadani
- TB-HIV Research Center, Faculty of Medicine, Universitas Padjadjaran, Nijmegen, The Netherlands
| | - Fitri Utama
- TB-HIV Research Center, Faculty of Medicine, Universitas Padjadjaran, Nijmegen, The Netherlands
| | - Rachel F Hannaway
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Agnes Indrati
- Department of Clinical Pathology, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin Hospital, Bandung, Indonesia
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, The Netherlands.,Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Romania
| | - Katrina Sharples
- Department of Mathematics and Statistics, Department of Preventive and Social Medicine, University of Otago, Dunedin, New Zealand
| | - Philip C Hill
- Centre for International Health, Department of Preventive and Social Medicine, University of Otago, Dunedin, New Zealand
| | - James E Ussher
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, The Netherlands
| |
Collapse
|
112
|
Franco AR, Peri F. Developing New Anti-Tuberculosis Vaccines: Focus on Adjuvants. Cells 2021; 10:cells10010078. [PMID: 33466444 PMCID: PMC7824815 DOI: 10.3390/cells10010078] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb) that sits in the top 10 leading causes of death in the world today and is the current leading cause of death among infectious diseases. Although there is a licensed vaccine against TB, the Mycobacterium bovis bacilli Calmette–Guérin (BCG) vaccine, it has several limitations, namely its high variability of efficacy in the population and low protection against pulmonary tuberculosis. New vaccines for TB are needed. The World Health Organization (WHO) considers the development and implementation of new TB vaccines to be a priority. Subunit vaccines are promising candidates since they can overcome safety concerns and optimize antigen targeting. Nevertheless, these vaccines need adjuvants in their formulation in order to increase immunogenicity, decrease the needed antigen dose, ensure a targeted delivery and optimize the antigens delivery and interaction with the immune cells. This review aims to focus on adjuvants being used in new formulations of TB vaccines, namely candidates already in clinical trials and others in preclinical development. Although no correlates of protection are defined, most research lines in the field of TB vaccination focus on T-helper 1 (Th1) type of response, namely polyfunctional CD4+ cells expressing simultaneously IFN-γ, TNF-α, and IL-2 cytokines, and also Th17 responses. Accordingly, most of the adjuvants reviewed here are able to promote such responses. In the future, it might be advantageous to consider a wider array of immune parameters to better understand the role of adjuvants in TB immunity and establish correlates of protection.
Collapse
|
113
|
Micoli F, Bagnoli F, Rappuoli R, Serruto D. The role of vaccines in combatting antimicrobial resistance. Nat Rev Microbiol 2021; 19:287-302. [PMID: 33542518 PMCID: PMC7861009 DOI: 10.1038/s41579-020-00506-3] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 01/29/2023]
Abstract
The use of antibiotics has enabled the successful treatment of bacterial infections, saving the lives and improving the health of many patients worldwide. However, the emergence and spread of antimicrobial resistance (AMR) has been highlighted as a global threat by different health organizations, and pathogens resistant to antimicrobials cause substantial morbidity and death. As resistance to multiple drugs increases, novel and effective therapies as well as prevention strategies are needed. In this Review, we discuss evidence that vaccines can have a major role in fighting AMR. Vaccines are used prophylactically, decreasing the number of infectious disease cases, and thus antibiotic use and the emergence and spread of AMR. We also describe the current state of development of vaccines against resistant bacterial pathogens that cause a substantial disease burden both in high-income countries and in low- and medium-income countries, discuss possible obstacles that hinder progress in vaccine development and speculate on the impact of next-generation vaccines against bacterial infectious diseases on AMR.
Collapse
Affiliation(s)
- Francesca Micoli
- grid.425088.3GSK Vaccines Institute for Global Health, Siena, Italy
| | | | | | | |
Collapse
|
114
|
Chen T, Blanc C, Liu Y, Ishida E, Singer S, Xu J, Joe M, Jenny-Avital ER, Chan J, Lowary TL, Achkar JM. Capsular glycan recognition provides antibody-mediated immunity against tuberculosis. J Clin Invest 2020; 130:1808-1822. [PMID: 31935198 DOI: 10.1172/jci128459] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 01/03/2020] [Indexed: 12/16/2022] Open
Abstract
A better understanding of all immune components involved in protecting against Mycobacterium tuberculosis infection is urgently needed to inform strategies for novel immunotherapy and tuberculosis (TB) vaccine development. Although cell-mediated immunity is critical, increasing evidence supports that antibodies also have a protective role against TB. Yet knowledge of protective antigens is limited. Analyzing sera from 97 US immigrants at various stages of M. tuberculosis infection, we showed protective in vitro and in vivo efficacy of polyclonal IgG against the M. tuberculosis capsular polysaccharide arabinomannan (AM). Using recently developed glycan arrays, we established that anti-AM IgG induced in natural infection is highly heterogeneous in its binding specificity and differs in both its reactivity to oligosaccharide motifs within AM and its functions in bacillus Calmette-Guérin vaccination and/or in controlled (latent) versus uncontrolled (TB) M. tuberculosis infection. We showed that anti-AM IgG from asymptomatic but not from diseased individuals was protective and provided data suggesting a potential role of IgG2 and specific AM oligosaccharides. Filling a gap in the current knowledge of protective antigens in humans, our data support the key role of the M. tuberculosis surface glycan AM and suggest the importance of targeting specific glycan epitopes within AM in antibody-mediated immunity against TB.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Caroline Blanc
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yanyan Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Elise Ishida
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sarah Singer
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jiayong Xu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maju Joe
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - John Chan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Todd L Lowary
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jacqueline M Achkar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
115
|
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: 23] [Impact Index Per Article: 5.8] [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
|
116
|
Martinot AJ, Blass E, Yu J, Aid M, Mahrokhian SH, Cohen SB, Plumlee CR, Larocca RA, Siddiqi N, Wakabayashi S, Gardner M, Audette R, Devorak A, Urdahl KB, Rubin EJ, Barouch DH. Protective efficacy of an attenuated Mtb ΔLprG vaccine in mice. PLoS Pathog 2020; 16:e1009096. [PMID: 33315936 PMCID: PMC7769599 DOI: 10.1371/journal.ppat.1009096] [Citation(s) in RCA: 9] [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: 06/07/2020] [Revised: 12/28/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
Bacille Calmette-Guerin (BCG), an attenuated whole cell vaccine based on Mycobacterium bovis, is the only licensed vaccine against Mycobacterium tuberculosis (Mtb), but its efficacy is suboptimal and it fails to protect against pulmonary tuberculosis. We previously reported that Mtb lacking the virulence genes lprG and rv1410c (ΔLprG) was highly attenuated in immune deficient mice. In this study, we show that attenuated ΔLprG Mtb protects C57BL/6J, Balb/cJ, and C3HeB/FeJ mice against Mtb challenge and is as attenuated as BCG in SCID mice. In C3HeB/FeJ mice, ΔLprG vaccination resulted in innate peripheral cytokine production and induced high polyclonal PPD-specific cytokine-secreting CD4+ T lymphocytes in peripheral blood. The ΔLprG vaccine afforded protective efficacy in the lungs of C3H/FeJ mice following both H37Rv and Erdman aerosolized Mtb challenges. Vaccine efficacy correlated with antigen-specific PD-1-negative CD4+ T lymphocytes as well as with serum IL-17 levels after vaccination. We hypothesize that induction of Th17 cells in lung is critical for vaccine protection, and we show a serum cytokine biomarker for IL-17 shortly after vaccination may predict protective efficacy. Many successful vaccines are based on attenuated human pathogens. The only licensed tuberculosis vaccine, BCG, is based on an attenuated version of live whole cell Mycobacterium bovis, the causative agent of tuberculosis (TB) in cattle. Advantages to using attenuated pathogens as vaccines include a broad antigen composition including proteins, lipids, carbohydrates and other molecules that can induce durable immune responses sometimes lasting decades. Here we test an attenuated Mycobacterium tuberculosis (Mtb), the causative agent of human TB, that lacks a key virulence factor as an alternative whole cell vaccine in mice. Attenuated Mtb lacking a key virulence protein, LprG, is immunogenic and protects mice against Mtb challenge. The LprG whole cell vaccine is protective in mice that develop lung pathology more similar to what is described in human TB and the LprG vaccine induces a key cytokine, IL-17, thought to be important for vaccine protection, in the peripheral blood early after vaccination. Together these data support the continued development of attenuated TB as a potential vaccine candidate. Furthermore our data suggests that serum IL-17 should be explored as a potential biomarker for vaccine efficacy in preclinical animal models.
Collapse
Affiliation(s)
- Amanda J. Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Infectious Diseases and Global Health, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shant H. Mahrokhian
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sara B. Cohen
- Department of Immunology, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Courtney R. Plumlee
- Department of Immunology, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Rafael A. Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Noman Siddiqi
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Shoko Wakabayashi
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Michelle Gardner
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Rebecca Audette
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Anne Devorak
- Department of Infectious Diseases and Global Health, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Kevin B. Urdahl
- Department of Immunology, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Departments of Pediatrics and Immunology, University of Washington, Seattle, Washington, United States of America
| | - Eric J. Rubin
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
117
|
Day TA, Penn-Nicholson A, Luabeya AKK, Fiore-Gartland A, Du Plessis N, Loxton AG, Vergara J, Rolf TA, Reid TD, Toefy A, Shenje J, Geldenhuys H, Tameris M, Mabwe S, Bilek N, Bekker LG, Diacon A, Walzl G, Ashman J, Frevol A, Sagawa ZK, Lindestam Arlehamn C, Sette A, Reed SG, Coler RN, Scriba TJ, Hatherill M. Safety and immunogenicity of the adjunct therapeutic vaccine ID93 + GLA-SE in adults who have completed treatment for tuberculosis: a randomised, double-blind, placebo-controlled, phase 2a trial. THE LANCET RESPIRATORY MEDICINE 2020; 9:373-386. [PMID: 33306991 DOI: 10.1016/s2213-2600(20)30319-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 06/01/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND A therapeutic vaccine that prevents recurrent tuberculosis would be a major advance in the development of shorter treatment regimens. We aimed to assess the safety and immunogenicity of the ID93 + GLA-SE vaccine at various doses and injection schedules in patients with previously treated tuberculosis. METHODS This randomised, double-blind, placebo-controlled, phase 2a trial was conducted at three clinical sites near Cape Town, South Africa. Patients were recruited at local clinics after receiving 4 months of tuberculosis treatment, and screened for eligibility after providing written informed consent. Participants were aged 18-60 years, BCG-vaccinated, HIV-uninfected, and diagnosed with drug-sensitive pulmonary tuberculosis. Eligible patients had completed standard treatment for pulmonary tuberculosis in the past 28 days. Participants were enrolled after completing standard treatment and randomly assigned sequentially to receive vaccine or placebo in three cohorts: 2 μg intramuscular ID93 + 2 μg GLA-SE on days 0 and 56 (cohort 1); 10 μg ID93 + 2 μg GLA-SE on days 0 and 56 (cohort 2); 2 μg ID93 + 5 μg GLA-SE on days 0 and 56 and placebo on day 28 (cohort 3); 2 μg ID93 + 5 μg GLA-SE on days 0, 28, and 56 (cohort 3); or placebo on days 0 and 56 (cohorts 1 and 2), with the placebo group for cohort 3 receiving an additional injection on day 28. Randomisation was in a ratio of 3:1 for ID93 + GLA-SE and saline placebo in cohorts 1 and 2, and in a ratio of 3:3:1 for (2 ×) ID93 + GLA-SE, (3 ×) ID93 + GLA-SE, and placebo in cohort 3. The primary outcomes were safety and immunogenicity (vaccine-specific antibody response and T-cell response). For the safety outcome, participants were observed for 30 min after each injection, injection site reactions and systemic adverse events were monitored until day 84, and serious adverse events and adverse events of special interest were monitored for 6 months after the last injection. Vaccine-specific antibody responses were measured by serum ELISA, and T-cell responses after stimulation with vaccine antigens were measured in cryopreserved peripheral blood mononuclear cells specimens using intracellular cytokine staining followed by flow cytometry. This study is registered with ClinicalTrials.gov, number NCT02465216. FINDINGS Between June 17, 2015, and May 30, 2016, we assessed 177 patients for inclusion. 61 eligible patients were randomly assigned to receive: saline placebo (n=5) or (2 ×) 2 μg ID93 + 2 μg GLA-SE (n=15) on days 0 and 56 (cohort 1); saline placebo (n=2) or (2 ×) 10 μg ID93 + 2 μg GLA-SE (n=5) on days 0 and 56 (cohort 2); saline placebo (n=5) on days 0, 28 and 56, or 2 μg ID93 + 5 μg GLA-SE (n=15) on days 0 and 56 and placebo injection on day 28, or (3 ×) 2 μg ID93 + 5 μg GLA-SE (n=14) on days 0, 28, and 56 (cohort 3). ID93 + GLA-SE induced robust and durable antibody responses and specific, polyfunctional CD4 T-cell responses to vaccine antigens. Two injections of the 2 μg ID93 + 5 μg GLA-SE dose induced antigen-specific IgG and CD4 T-cell responses that were significantly higher than those with placebo and persisted for the 6-month study duration. Mild to moderate injection site pain was reported after vaccination across all dose combinations, and induration and erythema in patients given 2 μg ID93 + 5 μg GLA-SE in two or three doses. One participant had grade 3 erythema and induration at the injection site. No vaccine-related serious adverse events were observed. INTERPRETATION Vaccination with ID93 + GLA-SE was safe and immunogenic for all tested regimens. These data support further evaluation of ID93 + GLA-SE in therapeutic vaccination strategies to improve tuberculosis treatment outcomes. FUNDING Wellcome Trust (102028/Z/13/Z).
Collapse
Affiliation(s)
- Tracey A Day
- Infectious Disease Research Institute, Seattle, WA, USA
| | - Adam Penn-Nicholson
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Angelique Kany Kany Luabeya
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nelita Du Plessis
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre G Loxton
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Julie Vergara
- Infectious Disease Research Institute, Seattle, WA, USA
| | - Tom A Rolf
- Infectious Disease Research Institute, Seattle, WA, USA
| | - Tim D Reid
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Asma Toefy
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Justin Shenje
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Hendrik Geldenhuys
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Michele Tameris
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Simbarashe Mabwe
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Linda-Gail Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | | | - Gerhard Walzl
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jill Ashman
- Infectious Disease Research Institute, Seattle, WA, USA
| | - Aude Frevol
- Infectious Disease Research Institute, Seattle, WA, USA
| | | | - Cecilia Lindestam Arlehamn
- Center for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; PAI Life Sciences, Seattle, WA, USA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| | | |
Collapse
|
118
|
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: 1.0] [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
|
119
|
O'Hagan DT, Lodaya RN, Lofano G. The continued advance of vaccine adjuvants - 'we can work it out'. Semin Immunol 2020; 50:101426. [PMID: 33257234 DOI: 10.1016/j.smim.2020.101426] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022]
Abstract
In the last decade there have been some significant advances in vaccine adjuvants, particularly in relation to their inclusion in licensed products. This was proceeded by several decades in which such advances were very scarce, or entirely absent, but several novel adjuvants have now been included in licensed products, including in the US. These advances have relied upon several key technological insights that have emerged in this time period, which have finally allowed an in depth understanding of how adjuvants work. These advances include developments in systems biology approaches which allow the hypotheses first advanced in pre-clinical studies to be critically evaluated in human studies. This review highlights these recent advances, both in relation to the adjuvants themselves, but also the technologies that have enabled their successes. Moreover, we critically appraise what will come next, both in terms of new adjuvant molecules, and the technologies needed to allow them to succeed. We confidently predict that additional adjuvants will emerge in the coming years that will reach approval in licensed products, but that the components might differ significantly from those which are currently used. Gradually, the natural products that were originally used to build adjuvants, since they were readily available at the time of initial development, will come to be replaced by synthetic or biosynthetic materials, with more appealing attributes, including more reliable and robust supply, along with reduced heterogeneity. The recent advance in vaccine adjuvants is timely, given the need to create novel vaccines to deal with the COVID-19 pandemic. Although, we must ensure that the rigorous safety evaluations that allowed the current adjuvants to advance are not 'short-changed' in the push for new vaccines to meet the global challenge as quickly as possible, we must not jeopardize what we have achieved, by pushing less established technologies too quickly, if the data does not fully support it.
Collapse
Affiliation(s)
- Derek T O'Hagan
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA
| | - Rushit N Lodaya
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA
| | - Giuseppe Lofano
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA.
| |
Collapse
|
120
|
Yamaguchi F, Yoda H, Hiraiwa M, Shiratori Y, Onozaki S, Ito M, Kashima S, Kosuge M, Atarashi K, Cho H, Shimizu S, Fujishima A, Mase A, Osakabe Y, Funaki T, Inoue D, Yamazaki Y, Tateno H, Yokoe T, Shikama Y. Impact of the interferon-γ release assay and glomerular filtration rate on the estimation of active tuberculosis risk before bronchoscopic examinations: a retrospective pilot study. J Thorac Dis 2020; 12:5842-5849. [PMID: 33209416 PMCID: PMC7656403 DOI: 10.21037/jtd-19-3653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Bronchoscopic examinations are vital to diagnose pulmonary diseases. However, as coughing is triggered during and after the procedure, it is imperative to take measures against nosocomial infections, especially for airborne infections like tuberculosis (TB). The interferon-γ release assay (IGRA) has recently been established as a method to evaluate the infection status of TB. We aimed to ascertain the efficacy of IGRA and clinical findings in estimating the prevalence of active TB before bronchoscopy. Methods We obtained IGRA results from 136 inpatients using a QuantiFERON-TB Gold In-Tube test. Bronchoscopy samples were cultured in Mycobacteria Growth indicator tubes and 2% Ogawa solid medium. We evaluated the adjusted effects of multiple clinical variables on active TB status using a logistic regression model. In addition, multiple variables were converted into a decision tree to predict active TB. Results Five (3.7%) patients were diagnosed with culture-positive TB, two of whom were simultaneously diagnosed with non-small-cell lung carcinoma or small-cell lung carcinoma. The multivariate analysis suggested the probability of predicting active TB using the IGRA [odds ratio (OR), 72.7; 95% confidence interval (CI), 3.169-1668; P=0.007] and decreased estimated glomerular filtration rate (eGFR) (OR, 0.937; 95% CI, 0.882-0.996; P=0.038) in patients undergoing bronchoscopy. A decision tree validated the use of these two variables to predict active TB. Conclusions IGRA test results are useful for predicting active TB before bronchoscopy. This strategy could identify patients who require antibiotic therapy to prevent TB or who are in the active phase of TB.
Collapse
Affiliation(s)
- Fumihiro Yamaguchi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Haruka Yoda
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Mina Hiraiwa
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yo Shiratori
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Shota Onozaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Mari Ito
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Saori Kashima
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Miku Kosuge
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kenji Atarashi
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hidekazu Cho
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Shohei Shimizu
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Akira Fujishima
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Ayaka Mase
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yuki Osakabe
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Toshitaka Funaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Daisuke Inoue
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yohei Yamazaki
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hidetsugu Tateno
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Takuya Yokoe
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Yusuke Shikama
- Department of Respiratory Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| |
Collapse
|
121
|
Khawbung JL, Nath D, Chakraborty S. Drug resistant Tuberculosis: A review. Comp Immunol Microbiol Infect Dis 2020; 74:101574. [PMID: 33249329 DOI: 10.1016/j.cimid.2020.101574] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) was announced as a global emergency in 1993. There was an alarming counter attack of TB worldwide. However, when it was known that TB can be cured completely, the general public became ignorant towards the infection. The pathogenic organism Mycobacterium tuberculosis continuously evolved to resist the antagonist drugs. This has led to the outbreak of resistant strain that gave rise to "Multi Drug Resistant-Tuberculosis" and "Extensively Drug Resistant Tuberculosis" that can still be cured with a lower success rate. While the mechanism of resistance proceeds further, it ultimately causes unmanageable totally drug resistant TB (TDR-TB). Studying the molecular mechanisms underlying the resistance to drugs would help us grasp the genetics and pathophysiology of the disease. In this review, we present the molecular mechanisms behind Mycobacterium tolerance to drugs and their approach towards the development of multi-drug resistant, extremely drug resistant and totally drug resistant TB.
Collapse
Affiliation(s)
| | - Durbba Nath
- Department of Biotechnology, Assam University, Silchar, 788011, Assam, India
| | - Supriyo Chakraborty
- Department of Biotechnology, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
122
|
Gebreselassie N, Kasaeva T, Zignol M. A global strategy for tuberculosis research and innovation. Eur Respir J 2020; 56:56/5/2003539. [PMID: 33184100 DOI: 10.1183/13993003.03539-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/18/2020] [Indexed: 11/05/2022]
Affiliation(s)
| | - Tereza Kasaeva
- Global TB Programme, World Health Organization, Geneva, Switzerland
| | - Matteo Zignol
- Global TB Programme, World Health Organization, Geneva, Switzerland
| |
Collapse
|
123
|
Counoupas C, Ferrell KC, Ashhurst A, Bhattacharyya ND, Nagalingam G, Stewart EL, Feng CG, Petrovsky N, Britton WJ, Triccas JA. Mucosal delivery of a multistage subunit vaccine promotes development of lung-resident memory T cells and affords interleukin-17-dependent protection against pulmonary tuberculosis. NPJ Vaccines 2020; 5:105. [PMID: 33298977 PMCID: PMC7665186 DOI: 10.1038/s41541-020-00255-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/02/2020] [Indexed: 11/29/2022] Open
Abstract
The development of effective vaccines against bacterial lung infections requires the induction of protective, pathogen-specific immune responses without deleterious inflammation within the pulmonary environment. Here, we made use of a polysaccharide-adjuvanted vaccine approach to elicit resident pulmonary T cells to protect against aerosol Mycobacterium tuberculosis infection. Intratracheal administration of the multistage fusion protein CysVac2 and the delta-inulin adjuvant Advax™ (formulated with a TLR9 agonist) provided superior protection against aerosol M. tuberculosis infection in mice, compared to parenteral delivery. Surprisingly, removal of the TLR9 agonist did not impact vaccine protection despite a reduction in cytokine-secreting T cell subsets, particularly CD4+IFN-γ+IL-2+TNF+ multifunctional T cells. CysVac2/Advax-mediated protection was associated with the induction of lung-resident, antigen-specific memory CD4+ T cells that expressed IL-17 and RORγT, the master transcriptional regulator of Th17 differentiation. IL-17 was identified as a key mediator of vaccine efficacy, with blocking of IL-17 during M. tuberculosis challenge reducing phagocyte influx, suppressing priming of pathogen-specific CD4+ T cells in local lymph nodes and ablating vaccine-induced protection. These findings suggest that tuberculosis vaccines such as CysVac2/Advax that are capable of eliciting Th17 lung-resident memory T cells are promising candidates for progression to human trials.
Collapse
Affiliation(s)
- Claudio Counoupas
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
| | - Kia C Ferrell
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Anneliese Ashhurst
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Nayan D Bhattacharyya
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Gayathri Nagalingam
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Erica L Stewart
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale and Flinders University, Adelaide, Australia
| | - Carl G Feng
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale and Flinders University, Adelaide, Australia
| | - Warwick J Britton
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - James A Triccas
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
- Charles Perkins Centre and Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
124
|
De Mot L, Bechtold V, Bol V, Callegaro A, Coccia M, Essaghir A, Hasdemir D, Ulloa-Montoya F, Siena E, Smilde A, van den Berg RA, Didierlaurent AM, Burny W, van der Most RG. Transcriptional profiles of adjuvanted hepatitis B vaccines display variable interindividual homogeneity but a shared core signature. Sci Transl Med 2020; 12:12/569/eaay8618. [DOI: 10.1126/scitranslmed.aay8618] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/23/2020] [Indexed: 12/19/2022]
Abstract
The current routine use of adjuvants in human vaccines provides a strong incentive to increase our understanding of how adjuvants differ in their ability to stimulate innate immunity and consequently enhance vaccine immunogenicity. Here, we evaluated gene expression profiles in cells from whole blood elicited in naive subjects receiving the hepatitis B surface antigen formulated with different adjuvants. We identified a core innate gene signature emerging 1 day after the second vaccination and that was shared by the recipients of vaccines formulated with adjuvant systems AS01B, AS01E, or AS03. This core signature associated with the magnitude of the hepatitis B surface-specific antibody response and was characterized by positive regulation of genes associated with interferon-related responses or the innate cell compartment and by negative regulation of natural killer cell–associated genes. Analysis at the individual subject level revealed that the higher immunogenicity of AS01B-adjuvanted vaccine was linked to its ability to induce this signature in most vaccinees even after the first vaccination. Therefore, our data suggest that adjuvanticity is not strictly defined by the nature of the receptors or signaling pathways it activates but by the ability of the adjuvant to consistently induce a core inflammatory signature across individuals.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dicle Hasdemir
- Bioinformatics Laboratory, University of Amsterdam, 1012 WX Amsterdam, Netherlands
- Biosystems Data Analysis Group, University of Amsterdam, 1012 WX Amsterdam, Netherlands
| | | | | | - Age Smilde
- Biosystems Data Analysis Group, University of Amsterdam, 1012 WX Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
125
|
Clemmensen HS, Knudsen NPH, Billeskov R, Rosenkrands I, Jungersen G, Aagaard C, Andersen P, Mortensen R. Rescuing ESAT-6 Specific CD4 T Cells From Terminal Differentiation Is Critical for Long-Term Control of Murine Mtb Infection. Front Immunol 2020; 11:585359. [PMID: 33240275 PMCID: PMC7677256 DOI: 10.3389/fimmu.2020.585359] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/12/2020] [Indexed: 12/25/2022] Open
Abstract
In most cases, Mycobacterium tuberculosis (Mtb) causes life-long chronic infections, which poses unique challenges for the immune system. Most of the current tuberculosis (TB) subunit vaccines incorporate immunodominant antigens and at this point, it is poorly understood how the CD4 T cell subsets recognizing these antigens are affected during long-term infection. Very little is known about the requirements for sustainable vaccine protection against TB. To explore this, we screened 62 human-recognized Mtb antigens during chronic murine Mtb infection and identified the four most immunodominant antigens in this setting (MPT70, Rv3020c, and Rv3019c and ESAT-6). Combined into a subunit vaccine, this fusion protein induced robust protection both in a standard short-term model and in a long-term infection model where immunity from BCG waned. Importantly, replacement of ESAT-6 with another ESAT-6-family antigen, Rv1198, led to similar short-term protection but a complete loss of bacterial control during chronic infection. This observation was further underscored, as the ESAT-6 containing vaccine mediated sustainable protection in a model of post-exposure vaccination, where the ESAT-6-replacement vaccine did not. An individual comparison of the CD4 T cell responses during Mtb infection revealed that ESAT-6-specific T cells were more terminally differentiated than the other immunodominant antigens and immunization with the ESAT-6 containing vaccine led to substantially greater reduction in the overall T cell differentiation status. Our data therefore associates long-term bacterial control with the ability of a vaccine to rescue infection-driven CD4T cell differentiation and future TB antigen discovery programs should focus on identifying antigens with the highest accompanying T cell differentiation, like ESAT-6. This also highlights the importance of long-term readouts in both preclinical and clinical studies with TB vaccines.
Collapse
Affiliation(s)
- Helena Strand Clemmensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.,Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Gregers Jungersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.,Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Claus Aagaard
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
126
|
Safety and Immunogenicity of the GamTBvac, the Recombinant Subunit Tuberculosis Vaccine Candidate: A Phase II, Multi-Center, Double-Blind, Randomized, Placebo-Controlled Study. Vaccines (Basel) 2020; 8:vaccines8040652. [PMID: 33153191 PMCID: PMC7712213 DOI: 10.3390/vaccines8040652] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 01/21/2023] Open
Abstract
GamTBvac is a candidate tuberculosis vaccine with two fusion proteins, containing Ag85a, ESAT6, CFP10, and a dextran-binding domain (DBD). Phase II of a double-blind, randomized, multicenter, placebo-controlled study in parallel groups in healthy adults to evaluate the safety and immunogenicity of GamTBvac in 180 previously-vaccinated with Bacillus Calmette-Guérin vaccine (BCG) healthy volunteers without Mycobacterium tuberculosis (MTB) infection was conducted. The dose (0.5 mL) of either the study drug or a placebo was administered subcutaneously twice with an 8-week interval. At eight timepoints from 14 to 150 days, whole blood and sera were assayed. Antigen-specific T-cell responses were measured by an in-house interferon-gamma release assay (IGRA-test), the QuantiFERON (QTF) test, and intracellular cytokine staining (ICS). For antibody response detection, the bead-based multiplex immunoassay (MIA) was applied. The vaccine confirmed an acceptable safety profile previously shown in a first-in-human clinical study. After stimulation with both fusions, the highest median level of INF-γ was detected on day 21. The GamTBvac vaccine induced antigen-specific interferon-gamma release, Th1 cytokine-expressing CD4+ T-cells, and IgG responses and results support further clinical testing of GamTBvac.
Collapse
|
127
|
Abstract
Vaccines save millions of lives from infectious diseases caused by viruses and bacteria. As the world awaits safe and effective COVID-19 vaccines, we celebrate the progresses made and highlight challenges ahead in vaccines and the science behind them.
Collapse
|
128
|
Tsujimura Y, Shiogama Y, Soma S, Okamura T, Takano J, Urano E, Murakata Y, Kawano A, Yamakawa N, Asaka MN, Matsuo K, Yasutomi Y. Vaccination with Intradermal Bacillus Calmette-Guérin Provides Robust Protection against Extrapulmonary Tuberculosis but Not Pulmonary Infection in Cynomolgus Macaques. THE JOURNAL OF IMMUNOLOGY 2020; 205:3023-3036. [PMID: 33097574 DOI: 10.4049/jimmunol.2000386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/22/2020] [Indexed: 01/16/2023]
Abstract
Recently, the efficacy of Mycobacterium bovis bacillus Calmette-Guérin (BCG) vaccination is being reassessed in accordance with the achievements of clinical tuberculosis (TB) vaccine research. However, the mechanisms ultimately determining the success or failure of BCG vaccination to prevent pulmonary TB remain poorly understood. In this study, we analyzed the protective effects of intradermal BCG vaccination by using specific pathogen-free cynomolgus macaques of Asian origin that were intradermally vaccinated with BCG (Tokyo strain) followed by Mycobacterium tuberculosis (Erdman strain) infection. Intradermal BCG administration generated TB Ag-specific multifunctional CD4 T cell responses in peripheral blood and bronchoalveolar lavage and almost completely protected against the development of TB pathogenesis with aggravation of clinical parameters and high levels of bacterial burdens in extrapulmonary organs. However, interestingly, there were no differences in bacterial quantitation and pathology of extensive granulomas in the lungs between BCG-vaccinated monkeys and control animals. These results indicated that the changes in clinical parameters, immunological responses, and quantitative gross pathology that are used routinely to determine the efficacy of TB vaccines in nonhuman primate models might not correlate with the bacterial burden and histopathological score in the lung as measured in this study.
Collapse
Affiliation(s)
- Yusuke Tsujimura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Yumiko Shiogama
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Shogo Soma
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan.,Department of Immunoregulation, Mie University Graduate School of Medicine, 514-8507 Tsu, Mie, Japan; and
| | - Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Junichiro Takano
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Emiko Urano
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Yoshiko Murakata
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Akira Kawano
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan.,Department of Immunoregulation, Mie University Graduate School of Medicine, 514-8507 Tsu, Mie, Japan; and.,Research and Development Department, Japan BCG Laboratory, 204-0022 Kiyose, Tokyo, Japan
| | - Natsuko Yamakawa
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Masamitsu N Asaka
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan
| | - Kazuhiro Matsuo
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan.,Research and Development Department, Japan BCG Laboratory, 204-0022 Kiyose, Tokyo, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, 305-0843 Tsukuba, Ibaraki, Japan; .,Department of Immunoregulation, Mie University Graduate School of Medicine, 514-8507 Tsu, Mie, Japan; and
| |
Collapse
|
129
|
Heaton PM. Challenges of Developing Novel Vaccines With Particular Global Health Importance. Front Immunol 2020; 11:517290. [PMID: 33162972 PMCID: PMC7591467 DOI: 10.3389/fimmu.2020.517290] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 09/03/2020] [Indexed: 01/12/2023] Open
Abstract
Six of the top ten leading causes of death in low resource settings could potentially be prevented by vaccination. Development of vaccines for individuals in these populations is difficult because of the biological complexity of the prevalent pathogens and the challenges inherent to development of any vaccine. This review discusses those challenges and promising advances to address them and highlights recent progress in development of vaccines against several pathogens of interest.
Collapse
Affiliation(s)
- Penny M Heaton
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, United States
| |
Collapse
|
130
|
Ault RC, Headley CA, Hare AE, Carruthers BJ, Mejias A, Turner J. Blood RNA signatures predict recent tuberculosis exposure in mice, macaques and humans. Sci Rep 2020; 10:16873. [PMID: 33037303 PMCID: PMC7547102 DOI: 10.1038/s41598-020-73942-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/18/2020] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death due to a single infectious disease. Knowing when a person was infected with Mycobacterium tuberculosis (M.tb) is critical as recent infection is the strongest clinical risk factor for progression to TB disease in immunocompetent individuals. However, time since M.tb infection is challenging to determine in routine clinical practice. To define a biomarker for recent TB exposure, we determined whether gene expression patterns in blood RNA correlated with time since M.tb infection or exposure. First, we found RNA signatures that accurately discriminated early and late time periods after experimental infection in mice and cynomolgus macaques. Next, we found a 6-gene blood RNA signature that identified recently exposed individuals in two independent human cohorts, including adult household contacts of TB cases and adolescents who recently acquired M.tb infection. Our work supports the need for future longitudinal studies of recent TB contacts to determine whether biomarkers of recent infection can provide prognostic information of TB disease risk in individuals and help map recent transmission in communities.
Collapse
Affiliation(s)
- Russell C Ault
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH, USA
- Medical Scientist Training Program, Ohio State University, Columbus, OH, USA
| | - Colwyn A Headley
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH, USA
| | - Alexander E Hare
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH, USA
- Medical Scientist Training Program, Ohio State University, Columbus, OH, USA
| | - Bridget J Carruthers
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Joanne Turner
- Texas Biomedical Research Institute, San Antonio, TX, USA.
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
131
|
Ullah I, Bibi S, Ul Haq I, Safia, Ullah K, Ge L, Shi X, Bin M, Niu H, Tian J, Zhu B. The Systematic Review and Meta-Analysis on the Immunogenicity and Safety of the Tuberculosis Subunit Vaccines M72/AS01 E and MVA85A. Front Immunol 2020; 11:1806. [PMID: 33133057 PMCID: PMC7578575 DOI: 10.3389/fimmu.2020.01806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023] Open
Abstract
Background: Tuberculosis (TB) is a severe infectious disease with devastating effects on global public health. No TB vaccine has yet been approved for use on latent TB infections and healthy adults. In this study, we performed a systematic review and meta-analysis to evaluate the immunogenicity and safety of the M72/AS01E and MVA85A subunit vaccines. The M72/AS01E is a novel peptide-based vaccine currently in progress, which may increase the protection level against TB infection. The MVA85A was a viral vector-based TB subunit vaccine being used in the clinical trials. The vaccines mentioned above have been studied in various phase I/II clinical trials. Immunogenicity and safety is the first consideration for TB vaccine development. Methods: The PubMed, Embase, and Cochrane Library databases were searched for published studies (until October 2019) to find out information on the M72/AS01E and MVA85A candidate vaccines. The meta-analysis was conducted by applying the standard methods and processes established by the Cochrane Collaboration. Results: Five eligible randomized clinical trials (RCTs) were selected for the meta-analysis of M72/AS01E candidate vaccines. The analysis revealed that the M72/AS01E subunit vaccine had an abundance of polyfunctional M72-specific CD4+ T cells [standardized mean difference (SMD) = 2.37] in the vaccine group versus the control group, the highest seropositivity rate [relative risk (RR) = 5.09]. The M72/AS01E vaccinated group were found to be at high risk of local injection site redness (RR = 2.64), headache (RR = 1.59), malaise (RR = 3.55), myalgia (RR = 2.27), fatigue (RR = 2.16), pain (RR = 3.99), swelling (RR = 5.09), and fever (RR = 2.04) compared to the control groups. The incidences of common adverse events of M72/AS01E were local injection site redness, headache, malaise, myalgia, fatigue, pain, swelling, fever, etc. Six eligible RCTs were selected for the meta-analysis on MVA85A candidate vaccines. The analysis revealed that the subunit vaccine MVA85A had a higher abundance of overall pooled proportion polyfunctional MVA85A-specific CD4+ T cells SMD = 2.41 in the vaccine group vs. the control group, with the highest seropositivity rate [estimation rate (ER) = 0.55]. The MVA85A vaccinated group were found to be at high risk of local injection site redness (ER = 0.55), headache (ER = 0.40), malaise (ER = 0.29), pain (ER = 0.54), myalgia (ER = 0.31), and fever (ER = 0.20). The incidences of common adverse events of MVA85A were local injection site redness, headache, malaise, pain, myalgia, fever, etc. Conclusion: The M72/AS01E and MVA85A vaccines against TB are safe and had immunogenicity in diverse clinical trials. The M72/AS01E and MVA85A vaccines are associated with a mild adverse reaction. The meta-analysis on immunogenicity and safety of M72/AS01E and MVA85A vaccines provides useful information for the evaluation of available subunit vaccines in the clinic.
Collapse
Affiliation(s)
- Inayat Ullah
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Shaheen Bibi
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China.,School of Life Science, Northwest Normal University, Lanzhou, China
| | - Ijaz Ul Haq
- College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, China
| | - Safia
- Pakistan Institute of Community Ophthalmology (PICO), Hayatabad Medical Complex, KMU, Peshawar, Pakistan
| | - Kifayat Ullah
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Long Ge
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xintong Shi
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Ma Bin
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Hongxia Niu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Jinhui Tian
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| |
Collapse
|
132
|
Harris RC, Sumner T, Knight GM, Zhang H, White RG. Potential impact of tuberculosis vaccines in China, South Africa, and India. Sci Transl Med 2020; 12:eaax4607. [PMID: 33028708 DOI: 10.1126/scitranslmed.aax4607] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 11/12/2019] [Accepted: 09/16/2020] [Indexed: 12/11/2022]
Abstract
More effective tuberculosis vaccines are needed to help reach World Health Organization tuberculosis elimination goals. Insufficient evidence exists on the potential impact of future tuberculosis vaccines with varying characteristics and in different epidemiological settings. To inform vaccine development decision making, we modeled the impact of hypothetical tuberculosis vaccines in three high-burden countries. We calibrated Mycobacterium tuberculosis (M.tb) transmission models to age-stratified demographic and epidemiological data from China, South Africa, and India. We varied vaccine efficacy to prevent infection or disease, effective in persons M.tb uninfected or infected, and duration of protection. We modeled routine early-adolescent vaccination and 10-yearly mass campaigns from 2025. We estimated median percentage population-level tuberculosis incidence rate reduction (IRR) in 2050 compared to a no new vaccine scenario. In all settings, results suggested vaccines preventing disease in M.tb-infected populations would have greatest impact by 2050 (10-year, 70% efficacy against disease, IRR 51%, 52%, and 54% in China, South Africa, and India, respectively). Vaccines preventing reinfection delivered lower potential impact (IRR 1, 12, and 17%). Intermediate impact was predicted for vaccines effective only in uninfected populations, if preventing infection (IRR 21, 37, and 50%) or disease (IRR 19, 36, and 51%), with greater impact in higher-transmission settings. Tuberculosis vaccines have the potential to deliver substantial population-level impact. For prioritizing impact by 2050, vaccine development should focus on preventing disease in M.tb-infected populations. Preventing infection or disease in uninfected populations may be useful in higher transmission settings. As vaccine impact depended on epidemiology, different development strategies may be required.
Collapse
Affiliation(s)
- Rebecca C Harris
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | - Tom Sumner
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Gwenan M Knight
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Hui Zhang
- Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Richard G White
- TB Modelling Group, TB Centre and Centre for the Mathematical Modelling of Infectious Diseases, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
133
|
Riou C, Du Bruyn E, Ruzive S, Goliath RT, Lindestam Arlehamn CS, Sette A, Sher A, Barber DL, Wilkinson RJ. Disease extent and anti-tubercular treatment response correlates with Mycobacterium tuberculosis-specific CD4 T-cell phenotype regardless of HIV-1 status. Clin Transl Immunology 2020; 9:e1176. [PMID: 33005414 PMCID: PMC7520805 DOI: 10.1002/cti2.1176] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives The development of non‐sputum‐based assays for tuberculosis (TB) diagnosis and treatment monitoring is a key priority. Recent data indicate that whole blood‐based assays to assess the phenotype of Mycobacterium tuberculosis (Mtb)‐specific CD4 T cells hold promise for this purpose and require further investigation in well‐characterised TB cohorts. In this study, we investigated the relationship between the phenotypic signature of Mtb‐specific CD4 responses, TB disease extent and treatment response. Methods Using flow cytometry, we measured the expression of phenotypic and functional markers (HLA‐DR, CD27, CD153, KLRG1, IL‐2, MIP‐1β, TNF‐α and IFN‐γ) on Mtb‐specific CD4 T‐cells in whole blood from 161 participants of varying TB and HIV status. TB disease extent was graded as a continuum using the Xpertct value, C‐reactive protein, Timika radiographic score and monocyte/lymphocyte ratio. Results The phenotypic profile of Mtb‐specific CD4 T cells pre‐anti‐tubercular treatment (ATT) strongly correlated with disease extent, irrespective of HIV status. ATT associated with major changes in the phenotype of Mtb‐specific CD4 T cells, with decreased expression of HLA‐DR and increased CD27 and CD153 expression. Principal component analysis showed an almost complete separation between latent TB infection (LTBI) and active TB (aTB) pre‐ATT groups, whereas the profile of the aTB post‐ATT group overlapped with the LTBI group. However, in patients experiencing treatment failure or relapse, no significant changes were observed in Mtb‐specific CD4 T‐cell phenotype pre‐ and post‐ATT. Conclusion Whole blood‐based assays of Mtb‐specific CD4 T‐cell activation and maturation markers can be used as non‐sputum‐based biomarkers of disease extent and treatment monitoring in TB, regardless of HIV‐1 status.
Collapse
Affiliation(s)
- Catherine Riou
- Wellcome Centre for Infectious Disease Research in Africa Institute of Infectious Disease and Molecular Medicine University of Cape Town Observatory South Africa.,Division of Immunology Department of Pathology University of Cape Town Observatory South Africa
| | - Elsa Du Bruyn
- Wellcome Centre for Infectious Disease Research in Africa Institute of Infectious Disease and Molecular Medicine University of Cape Town Observatory South Africa
| | - Sheena Ruzive
- Wellcome Centre for Infectious Disease Research in Africa Institute of Infectious Disease and Molecular Medicine University of Cape Town Observatory South Africa
| | - Rene T Goliath
- Wellcome Centre for Infectious Disease Research in Africa Institute of Infectious Disease and Molecular Medicine University of Cape Town Observatory South Africa
| | | | - Alessandro Sette
- Division of Vaccine Discovery La Jolla Institute for Immunology La Jolla CA USA.,Department of Medicine University of California San Diego La Jolla CA USA
| | - Alan Sher
- Immunobiology Section Laboratory of Parasitic Diseases National Institute of Allergy and Infectious Diseases National Institutes of Health Bethesda MD USA
| | - Daniel L Barber
- T Lymphocyte Biology Section Laboratory of Parasitic Diseases National Institute of Allergy and Infectious Diseases National Institutes of Health Bethesda MD USA
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa Institute of Infectious Disease and Molecular Medicine University of Cape Town Observatory South Africa.,Department of Infectious Diseases Imperial College London London UK.,Department of Medicine University of Cape Town Observatory South Africa.,The Francis Crick Institute London UK
| |
Collapse
|
134
|
Shi S, Liang Z, Sun B. Response to comment on: Vaccine adjuvants: Understanding the structure and mechanism of adjuvanticity. Vaccine 2020; 38:2759. [PMID: 32145881 DOI: 10.1016/j.vaccine.2020.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/05/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Shuting Shi
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Zhihui Liang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China.
| |
Collapse
|
135
|
Peters JS, Ismail N, Dippenaar A, Ma S, Sherman DR, Warren RM, Kana BD. Genetic Diversity in Mycobacterium tuberculosis Clinical Isolates and Resulting Outcomes of Tuberculosis Infection and Disease. Annu Rev Genet 2020; 54:511-537. [PMID: 32926793 DOI: 10.1146/annurev-genet-022820-085940] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tuberculosis claims more human lives than any other bacterial infectious disease and represents a clear and present danger to global health as new tools for vaccination, treatment, and interruption of transmission have been slow to emerge. Additionally, tuberculosis presents with notable clinical heterogeneity, which complicates diagnosis, treatment, and the establishment of nonrelapsing cure. How this heterogeneity is driven by the diversity ofclinical isolates of the causative agent, Mycobacterium tuberculosis, has recently garnered attention. Herein, we review advances in the understanding of how naturally occurring variation in clinical isolates affects transmissibility, pathogenesis, immune modulation, and drug resistance. We also summarize how specific changes in transcriptional responses can modulate infection or disease outcome, together with strain-specific effects on gene essentiality. Further understanding of how this diversity of M. tuberculosis isolates affects disease and treatment outcomes will enable the development of more effective therapeutic options and vaccines for this dreaded disease.
Collapse
Affiliation(s)
- Julian S Peters
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg 2000, South Africa; ,
| | - Nabila Ismail
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; ,
| | - Anzaan Dippenaar
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; , .,Family Medicine and Population Health (FAMPOP), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, 2000, Belgium;
| | - Shuyi Ma
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - David R Sherman
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Robin M Warren
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; ,
| | - Bavesh D Kana
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg 2000, South Africa; ,
| |
Collapse
|
136
|
Expanding the role of bacterial vaccines into life-course vaccination strategies and prevention of antimicrobial-resistant infections. NPJ Vaccines 2020; 5:84. [PMID: 32963814 PMCID: PMC7486369 DOI: 10.1038/s41541-020-00232-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/19/2020] [Indexed: 12/28/2022] Open
Abstract
A crisis in bacterial infections looms as ageing populations, increasing rates of bacteraemia and healthcare-associated infections converge with increasing antimicrobial resistance and a paucity of new antimicrobial classes. New initiatives are needed to develop bacterial vaccines for older adults in whom immune senescence plays a critical role. Novel vaccines require an expanded repertoire to prevent mucosal diseases such as pneumonia, skin and soft tissue infections and urinary tract infections that are major causes of morbidity and mortality in the elderly, and key drivers of antimicrobial resistance. This review considers the challenges inherent to the prevention of bacterial diseases, particularly mucosal infections caused by major priority bacterial pathogens against which current vaccines are sub-optimal. It has become clear that prevention of many lung, urinary tract and skin infections requires more than circulating antibodies. Induction of Th1/Th17 cellular responses with tissue-resident memory (Trm) cells homing to mucosal tissues may be a pre-requisite for success.
Collapse
|
137
|
Li Z, Zheng C, Terreni M, Tanzi L, Sollogoub M, Zhang Y. Novel Vaccine Candidates against Tuberculosis. Curr Med Chem 2020; 27:5095-5118. [DOI: 10.2174/0929867326666181126112124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Ranking above AIDS, Tuberculosis (TB) is the ninth leading cause of death affecting and
killing many individuals every year. Drugs’ efficacy is limited by a series of problems such as Multi-
Drug Resistance (MDR) and Extensively-Drug Resistance (XDR). Meanwhile, the only licensed vaccine
BCG (Bacillus Calmette-Guérin) existing for over 90 years is not effective enough. Consequently,
it is essential to develop novel vaccines for TB prevention and immunotherapy. This paper
provides an overall review of the TB prevalence, immune system response against TB and recent
progress of TB vaccine research and development. Several vaccines in clinical trials are described as
well as LAM-based candidates.
Collapse
Affiliation(s)
- Zhihao Li
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Changping Zheng
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Marco Terreni
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Lisa Tanzi
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Matthieu Sollogoub
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Yongmin Zhang
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
138
|
Preclinical Progress of Subunit and Live Attenuated Mycobacterium tuberculosis Vaccines: A Review following the First in Human Efficacy Trial. Pharmaceutics 2020; 12:pharmaceutics12090848. [PMID: 32899930 PMCID: PMC7559421 DOI: 10.3390/pharmaceutics12090848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/24/2022] Open
Abstract
Tuberculosis (TB) is the global leading cause of death from an infectious agent with approximately 10 million new cases of TB and 1.45 million deaths in 2018. Bacille Calmette-Guérin (BCG) remains the only approved vaccine for Mycobacterium tuberculosis (M. tb, causative agent of TB), however clinical studies have shown BCG has variable effectiveness ranging from 0–80% in adults. With 1.7 billion people latently infected, it is becoming clear that vaccine regimens aimed at both post-exposure and pre-exposure to M. tb will be crucial to end the TB epidemic. The two main strategies to improve or replace BCG are subunit and live attenuated vaccines. However, following the failure of the MVA85A phase IIb trial in 2013, more varied and innovative approaches are being developed. These include recombinant BCG strains, genetically attenuated M. tb and naturally attenuated mycobacteria strains, novel methods of immunogenic antigen discovery including for hypervirulent M. tb strains, improved antigen recognition and delivery strategies, and broader selection of viral vectors. This article reviews preclinical vaccine work in the last 5 years with focus on those tested against M. tb challenge in relevant animal models.
Collapse
|
139
|
Saralahti AK, Uusi-Mäkelä MIE, Niskanen MT, Rämet M. Integrating fish models in tuberculosis vaccine development. Dis Model Mech 2020; 13:13/8/dmm045716. [PMID: 32859577 PMCID: PMC7473647 DOI: 10.1242/dmm.045716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis is a chronic infection by Mycobacterium tuberculosis that results in over 1.5 million deaths worldwide each year. Currently, there is only one vaccine against tuberculosis, the Bacillus Calmette–Guérin (BCG) vaccine. Despite widespread vaccination programmes, over 10 million new M. tuberculosis infections are diagnosed yearly, with almost half a million cases caused by antibiotic-resistant strains. Novel vaccination strategies concentrate mainly on replacing BCG or boosting its efficacy and depend on animal models that accurately recapitulate the human disease. However, efforts to produce new vaccines against an M. tuberculosis infection have encountered several challenges, including the complexity of M. tuberculosis pathogenesis and limited knowledge of the protective immune responses. The preclinical evaluation of novel tuberculosis vaccine candidates is also hampered by the lack of an appropriate animal model that could accurately predict the protective effect of vaccines in humans. Here, we review the role of zebrafish (Danio rerio) and other fish models in the development of novel vaccines against tuberculosis and discuss how these models complement the more traditional mammalian models of tuberculosis. Summary: In this Review, we discuss how zebrafish (Danio rerio) and other fish models can complement the more traditional mammalian models in the development of novel vaccines against tuberculosis.
Collapse
Affiliation(s)
- Anni K Saralahti
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland
| | - Meri I E Uusi-Mäkelä
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland
| | - Mirja T Niskanen
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland .,Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland.,PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu FI-90014, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu FI-90029, Finland
| |
Collapse
|
140
|
Davids M, Pooran A, Hermann C, Mottay L, Thompson F, Cardenas J, Gu J, Koeuth T, Meldau R, Limberis J, Gina P, Srivastava S, Calder B, Esmail A, Tomasicchio M, Blackburn J, Gumbo T, Dheda K. A Human Lung Challenge Model to Evaluate the Safety and Immunogenicity of PPD and Live Bacillus Calmette-Guérin. Am J Respir Crit Care Med 2020; 201:1277-1291. [PMID: 31860339 DOI: 10.1164/rccm.201908-1580oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: A human model to better understand tuberculosis immunopathogenesis and facilitate vaccine development is urgently needed.Objectives: We evaluated the feasibility, safety, and immunogenicity of live bacillus Calmette-Guérin (BCG) in a lung-oriented controlled human infection model.Methods: We recruited 106 healthy South African participants with varying degrees of tuberculosis susceptibility. Live BCG, sterile PPD, and saline were bronchoscopically instilled into separate lung segments (n = 65). A control group (n = 34) underwent a single bronchoscopy without challenge. The primary outcome was safety. Cellular and antibody immune signatures were identified in BAL before and 3 days after challenge using flow cytometry, ELISA, RNA sequencing, and mass spectrometry.Measurements and Main Results: The frequency of adverse events was low (9.4%; n = 10), similar in the challenge versus control groups (P = 0.8), and all adverse events were mild and managed conservatively in an outpatient setting. The optimal PPD and BCG dose was 0.5 TU and 104 cfu, respectively, based on changes in BAL cellular profiles (P = 0.02) and antibody responses (P = 0.01) at incremental doses before versus after challenge. At 104 versus 103 cfu BCG, there was a significant increase in number of differentially expressed genes (367 vs. 3; P < 0.001) and dysregulated proteins (64 vs. 0; P < 0.001). Immune responses were highly setting specific (in vitro vs. in vivo) and compartment specific (BAL vs. blood) and localized to the challenged lung segments.Conclusions: A lung-oriented mycobacterial controlled human infection model using live BCG and PPD is feasible and safe. These data inform the study of tuberculosis immunopathogenesis and strategies for evaluation and development of tuberculosis vaccine candidates.
Collapse
Affiliation(s)
- Malika Davids
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Anil Pooran
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Clemens Hermann
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Lynelle Mottay
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Fawziyah Thompson
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Jacob Cardenas
- Baylor Institute for Immunology Research, Dallas, Texas; and
| | - Jinghua Gu
- Baylor Institute for Immunology Research, Dallas, Texas; and
| | - Thearith Koeuth
- Baylor Institute for Immunology Research, Dallas, Texas; and
| | - Richard Meldau
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Jason Limberis
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Phindile Gina
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | | | - Bridget Calder
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Aliasgar Esmail
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Michele Tomasicchio
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa
| | - Jonathan Blackburn
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Tawanda Gumbo
- Baylor Institute for Immunology Research, Dallas, Texas; and
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa.,South African Medical Research Council/University of Cape Town Centre for the Study of Antimicrobial Resistance, Cape Town, South Africa.,Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
141
|
Nutritional status positively impacts humoral immunity against its Mycobacterium tuberculosis, disease progression, and vaccine development. PLoS One 2020; 15:e0237062. [PMID: 32760105 PMCID: PMC7410285 DOI: 10.1371/journal.pone.0237062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 07/20/2020] [Indexed: 11/25/2022] Open
Abstract
Nutritional status contributes to the regulation of immune responses against pathogens, and malnutrition has been considered as a risk factor for tuberculosis (TB). Mycobacterium tuberculosis (Mtb), the causative agent of TB, can modulate host lipid metabolism and induce lipid accumulation in macrophages, where the bacilli adopt a dormant phenotype. In addition, serum lipid components play dual roles in the regulation of and protection from Mtb infection. We analyzed the relationship between nutritional status and the humoral immune response in TB patients. We found that serum HDL levels are positively correlated with the serum IgA specific for Mtb antigens. Analysis of the relationship between serum nutritional parameters and clinical parameters in TB patients showed that serum albumin and CRP levels were negatively correlated before treatment. We also observed reduced serum LDL levels in TB patients following treatment. These findings may provide insight into the role of serum lipids in host immune responses against Mtb infection. Furthermore, improving the nutritional status may enhance vaccination efficacy.
Collapse
|
142
|
Katelaris AL, Jackson C, Southern J, Gupta RK, Drobniewski F, Lalvani A, Lipman M, Mangtani P, Abubakar I. Effectiveness of BCG Vaccination Against Mycobacterium tuberculosis Infection in Adults: A Cross-sectional Analysis of a UK-Based Cohort. J Infect Dis 2020; 221:146-155. [PMID: 31504674 DOI: 10.1093/infdis/jiz430] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND BCG appears to reduce acquisition of Mycobacterium tuberculosis infection in children, measured using interferon-gamma release assays (IGRAs). We explored whether BCG vaccination continues to be associated with decreased prevalence of M. tuberculosis infection in adults. METHODS We conducted a cross-sectional analysis of data from adult contacts of tuberculosis cases participating in a UK cohort study. Vaccine effectiveness (VE) of BCG, ascertained based on presence of a scar or vaccination history, against latent tuberculosis infection (LTBI), measured via IGRA, was assessed using multivariable logistic regression. The effects of age at BCG and time since vaccination were also explored. RESULTS Of 3453 recent tuberculosis contacts, 27.5% had LTBI. There was strong evidence of an association between BCG and LTBI (adjusted odds ratio = 0.70; 95% confidence interval, .56-.87; P = .0017) yielding a VE of 30%. VE declined with time since vaccination but there was evidence that LTBI prevalence was lower amongst vaccinated individuals even >20 years after vaccination, compared with nonvaccinated participants. CONCLUSIONS BCG is associated with lower prevalence of LTBI in adult contacts of tuberculosis. These results contribute to growing evidence that suggests BCG may protect against M. tuberculosis infection as well as disease. This has implications for immunization programs, vaccine development, and tuberculosis control efforts worldwide. CLINICAL TRIALS REGISTRATION NCT01162265.
Collapse
Affiliation(s)
- Anthea L Katelaris
- Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine
| | | | - Jo Southern
- National Infection Service, Public Health England
| | - Rishi K Gupta
- Institute for Global Health, University College London
| | | | - Ajit Lalvani
- National Institute for Health Research Health Protection Research Unit in Respiratory Infections, Imperial College, London, United Kingdom
| | - Marc Lipman
- University College London Respiratory, Division of Medicine, University College London
| | - Punam Mangtani
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine
| | | |
Collapse
|
143
|
Li J, Zhao A, Tang J, Wang G, Shi Y, Zhan L, Qin C. Tuberculosis vaccine development: from classic to clinical candidates. Eur J Clin Microbiol Infect Dis 2020; 39:1405-1425. [PMID: 32060754 PMCID: PMC7223099 DOI: 10.1007/s10096-020-03843-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Bacillus Calmette-Guérin (BCG) has been in use for nearly 100 years and is the only licensed TB vaccine. While BCG provides protection against disseminated TB in infants, its protection against adult pulmonary tuberculosis (PTB) is variable. To achieve the ambitious goal of eradicating TB worldwide by 2050, there is an urgent need to develop novel TB vaccines. Currently, there are more than a dozen novel TB vaccines including prophylactic and therapeutic at different stages of clinical research. This literature review provides an overview of the clinical status of candidate TB vaccines and discusses the challenges and future development trends of novel TB vaccine research in combination with the efficacy of evaluation of TB vaccines, provides insight for the development of safer and more efficient vaccines, and may inspire new ideas for the prevention of TB.
Collapse
Affiliation(s)
- Junli Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Aihua Zhao
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Jun Tang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Guozhi Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Yanan Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Lingjun Zhan
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| |
Collapse
|
144
|
Scriba TJ, Netea MG, Ginsberg AM. Key recent advances in TB vaccine development and understanding of protective immune responses against Mycobacterium tuberculosis. Semin Immunol 2020; 50:101431. [PMID: 33279383 PMCID: PMC7786643 DOI: 10.1016/j.smim.2020.101431] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/02/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
Tuberculosis is the leading infectious disease killer globally due to a single pathogen. Despite wide deployment of standard drug regimens, modern diagnostics and a vaccine (bacille Calmette Guerin, BCG), the global tuberculosis epidemic is inadequately controlled. Novel, effective vaccine(s) are a crucial element of the World Health Organization End TB Strategy. TB vaccine research and development has recently been catalysed by several factors, including a revised strategy focused first on preventing pulmonary TB in adolescents and adults who are the main source of transmission, and encouraging evaluations of novel efficacy endpoints. Renewed enthusiasm for TB vaccine research has also been stimulated by recent preclinical and clinical advancements. These include new insights into underlying protective immune responses, including potential roles for 'trained' innate immunity and Th1/Th17 CD4+ (and CD8+) T cells. The field has been further reinvigorated by two positive proof of concept efficacy trials: one evaluating a potential new use of BCG in preventing high risk populations from sustained Mycobacterium tuberculosis infection and the second evaluating a novel, adjuvanted, recombinant protein vaccine candidate (M72/AS01E) for prevention of disease in adults already infected. Fourteen additional candidates are currently in various phases of clinical evaluation and multiple approaches to next generation vaccines are in discovery and preclinical development. The two positive efficacy trials and recent studies in nonhuman primates have enabled the first opportunities to discover candidate vaccine-induced correlates of protection, an effort being undertaken by a broad research consortium.
Collapse
Affiliation(s)
- Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Geert Grooteplein 8, 6525 GA Nijmegen, the Netherlands; Department of Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany.
| | - Ann M Ginsberg
- Bill & Melinda Gates Foundation, Division of Global Health, Washington DC, United States.
| |
Collapse
|
145
|
Niskanen M, Myllymäki H, Rämet M. DNA vaccination with the Mycobacterium marinum MMAR_4110 antigen inhibits reactivation of a latent mycobacterial infection in the adult Zebrafish. Vaccine 2020; 38:5685-5694. [PMID: 32624250 DOI: 10.1016/j.vaccine.2020.06.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/03/2020] [Accepted: 06/18/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Tuberculosis is a major challenge for health care, as options for its treatment and prevention are limited. Therefore, novel approaches, such as DNA vaccination, to both prevent primary infections and the reactivation of latent infections need to be developed. A Mycobacterium marinum infection in adult zebrafish (Danio rerio) recapitulates features of the human Mycobacterium tuberculosis infection, providing a convenient preclinical animal model for studying tuberculosis. METHODS Hypoxic M. marinum cultures were produced with the Wayne model, and further reaerated to replicate the in vivo reactivation in vitro. Expression levels of M. marinum genes were studied with mRNA sequencing from exponentially growing bacteria, anaerobic cultures and at 2 and 12 h after reaeration. Seven reactivation-associated genes were selected for further studies, where their antigen potentiality as DNA-vaccines to prevent reactivation of a latent mycobacterial infection was investigated in the adult zebrafish model. The Mann-Whitney test was used to evaluate differences in bacterial counts between the groups. RESULTS The mRNA sequencing data showed that, seven M. marinum genes, MMAR_0444, MMAR_0514, MMAR_0552, MMAR_0641, MMAR_1093, MMAR_4110 and MMAR_4524, were upregulated during reactivation when compared to both dormant and logarithmic growing bacteria. Four different MMAR_4110 antigens prevented the reactivation of a latent mycobacterial infection in the adult zebrafish. CONCLUSION This study provides novel information about reactivation-related M. marinum genes. One of the antigens, MMAR_4110, inhibited the reactivation of a latent M. marinum infection in zebrafish, implicating that the characterized genes could be potential targets for further vaccine and drug development against mycobacterial diseases.
Collapse
Affiliation(s)
- Mirja Niskanen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Henna Myllymäki
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Rämet
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland; PEDEGO Research Unit and Medical Research Centre, University of Oulu, Finland.
| |
Collapse
|
146
|
Ogongo P, Steyn AJ, Karim F, Dullabh KJ, Awala I, Madansein R, Leslie A, Behar SM. Differential skewing of donor-unrestricted and γδ T cell repertoires in tuberculosis-infected human lungs. J Clin Invest 2020; 130:214-230. [PMID: 31763997 PMCID: PMC6934215 DOI: 10.1172/jci130711] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Unconventional T cells that recognize mycobacterial antigens are of great interest as potential vaccine targets against tuberculosis (TB). This includes donor-unrestricted T cells (DURTs), such as mucosa-associated invariant T cells (MAITs), CD1-restricted T cells, and γδ T cells. We exploited the distinctive nature of DURTs and γδ T cell receptors (TCRs) to investigate the involvement of these T cells during TB in the human lung by global TCR sequencing. Making use of surgical lung resections, we investigated the distribution, frequency, and characteristics of TCRs in lung tissue and matched blood from individuals infected with TB. Despite depletion of MAITs and certain CD1-restricted T cells from the blood, we found that the DURT repertoire was well preserved in the lungs, irrespective of disease status or HIV coinfection. The TCRδ repertoire, in contrast, was highly skewed in the lungs, where it was dominated by Vδ1 and distinguished by highly localized clonal expansions, consistent with the nonrecirculating lung-resident γδ T cell population. These data show that repertoire sequencing is a powerful tool for tracking T cell subsets during disease.
Collapse
Affiliation(s)
- Paul Ogongo
- Africa Health Research Institute and.,School of Laboratory Medicine, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.,Institute of Primate Research, National Museums of Kenya, Nairobi, Kenya
| | | | | | - Kaylesh J Dullabh
- Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Ismael Awala
- Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Rajhmun Madansein
- Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute and.,Department of Infection and Immunity, University College London, London, United Kingdom
| | - Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
147
|
Dalirfardouei R, Tafaghodi M, Meshkat Z, Najafi A, Gholoobi A, Nabavinia MS, Sajedifar S, Meshkat M, Badiee A, Ramezani M, Varasteh AR, Naderinasab M. A novel formulation of Mtb72F DNA vaccine for immunization against tuberculosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:826-832. [PMID: 32695300 PMCID: PMC7351443 DOI: 10.22038/ijbms.2020.41806.9881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective(s): Mycobacterium tuberculosis (M. tuberculosis), an intracellular pathogen, causes 1.5 million deaths globally. Bacilli Calmette-Guérin (BCG) is commonly administered to protect people against M. tuberculosis infection; however, there are some obstacles with this first-generation vaccine. DNA vaccines, the third generation vaccines, can induce cellular immune responses for tuberculosis (TB) protection. In this study, optimized DNA vaccine (pcDNA3.1-Mtb72F) entrapped in poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) was used to achieve higher immunogenicity. Materials and Methods: Plasmid Mtb72F was formulated in PLGA NPs using double emulsion method in the presence of TB10.4 and/or CpG as an adjuvant. Female BALB/c mice were immunized either with NP-encapsulated Mtb72F or naked Mtb72F with or without each adjuvant, using the BCG-prime DNA boost regimen. Results: These NPs were approximately 250 nm in diameter and the nucleic acid and protein encapsulation efficiency were 80% and 25%, respectively. The NPs smaller than 200 nm are able to promote cellular rather than humoral responses. The immunization with the formulation consisting of Mtb72F DNA vaccine and TB10.4 entrapped in PLGA NPs showed significant immunogenicity and induced predominantly interferon-ɣ (IFN-ɣ) production and higher INF-ɣ/interleukin-4 (IL-4) ratio in the cultured spleen cells supernatant. Conclusion: PLGA NPs loaded with Mtb72F DNA-based vaccine with TB10.4 could be considered as a promising candidate for vaccination against TB. These results represent an excellent initial step toward development of novel vaccine for TB protection.
Collapse
Affiliation(s)
- Razieh Dalirfardouei
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Adel Najafi
- Laboratory Division, Fatemieh Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aida Gholoobi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Sadat Nabavinia
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | | - Ali Badiee
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdol-Reza Varasteh
- Immunobiochemistry Lab, Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboubeh Naderinasab
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
148
|
The potential impact of preventive therapy against tuberculosis in the WHO South-East Asian Region: a modelling approach. BMC Med 2020; 18:163. [PMID: 32684164 PMCID: PMC7369473 DOI: 10.1186/s12916-020-01651-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The prevention of tuberculosis (TB) is key for accelerating current, slow declines in TB burden. The 2018 World Health Organization (WHO) guidelines on eligibility for preventive therapy to treat latent TB infection (LTBI) include people living with human immunodeficiency virus (PLHIV), household contacts of TB patients including children, and those with clinical conditions including silicosis, dialysis, transplantation, etc. and other country-specific groups. We aimed to estimate the potential impact of full implementation of these guidelines in the WHO South-East Asian (SEA) Region, which bears the largest burden of TB and LTBI amongst the WHO regions. METHODS We developed mathematical models of TB transmission dynamics, calibrated individually to each of the 11 countries in the region. We modelled preventive therapy in the absence of other TB interventions. As an alternative comparator, reflecting ongoing developments in TB control in the region, we also simulated improvements in the treatment cascade for active TB, including private sector engagement and intensified case-finding. Relative to both scenarios, for each country in the region, we projected TB cases and deaths averted between 2020 and 2030, by full uptake of preventive therapy, defined as comprehensive coverage amongst eligible populations as per WHO guidelines, and assuming outcomes consistent with clinical trials. We also performed sensitivity analysis to illustrate impact under less-than-optimal conditions. RESULTS At the regional level, full uptake of preventive therapy amongst identified risk groups would reduce annual incidence rates in 2030 by 8.30% (95% CrI 6.48-10.83) relative to 2015, in the absence of any additional interventions. If implemented against a backdrop of improved TB treatment cascades, preventive therapy would achieve an incremental 6.93 percentage points (95% CrI 5.81-8.51) of reduction in annual incidence rates, compared to 2015. At the regional level, the numbers of individuals with latent TB infection that need to be treated to avert 1 TB case is 64 (95% CrI 55-74). Sensitivity analysis illustrates that results for impact are roughly proportional to 'effective coverage' (the product of actual coverage and effectiveness of the regimen). CONCLUSIONS Full implementation of WHO guidelines is important for ending TB in the SEA Region. Although future strategies will need to be expanded to the population level, to achieve large declines in TB incidence, the uptake of current tools can offer a valuable step in this direction.
Collapse
|
149
|
Rasheed OK, Ettenger G, Buhl C, Child R, Miller SM, Evans JT, Ryter KT. 6,6'-Aryl trehalose analogs as potential Mincle ligands. Bioorg Med Chem 2020; 28:115564. [PMID: 32616186 PMCID: PMC7372699 DOI: 10.1016/j.bmc.2020.115564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/27/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
6,6'-Aryl trehalose derivatives have been synthesized with a view towards identifying novel Th-17-inducing vaccine adjuvants based on the high affinity Mincle ligand Brartemicin. The initial structure-activity relationships of these novel trehalose-based compounds were investigated. All compounds have been evaluated for their ability to engage the Mincle receptor and induce a potential pro-Th17 cytokine profile from human peripheral blood mononuclear cells based on IL-6 production in human peripheral blood mononuclear cells. The preliminary biological characterization of the designed analogs presented in this paper should aid in the future design and testing of more affine ligands that may foster the discovery of novel adjuvants with improved pharmacological properties.
Collapse
Affiliation(s)
- Omer K Rasheed
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - George Ettenger
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Cassandra Buhl
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Robert Child
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Shannon M Miller
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Kendal T Ryter
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States.
| |
Collapse
|
150
|
Porcelli SA, Jacobs WR. Exacting Edward Jenner's revenge: The quest for a new tuberculosis vaccine. Sci Transl Med 2020; 11:11/490/eaax4219. [PMID: 31043569 PMCID: PMC6943913 DOI: 10.1126/scitranslmed.aax4219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/10/2019] [Indexed: 12/25/2022]
Abstract
Recent experimental and clinical work has reinvigorated the pursuit of a better tuberculosis vaccine.
Collapse
Affiliation(s)
- Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|