1
|
Zhang M, Ma J, Li M. Original Antigenic Sin in CD4+ T Cells. Immunology 2025; 175:165-179. [PMID: 40056013 DOI: 10.1111/imm.13916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 05/07/2025] Open
Abstract
Original antigenic sin (OAS) describes the phenomenon in which prior exposure to an antigen weakens the adaptive antibody response to a subsequent heterologous infection. This phenomenon can diminish the effectiveness of immunity acquired through vaccination or previous infections. We demonstrate that OAS arises because CD4+ T cell proliferation and regulation signals are antigen-nonspecific. Rapidly responding memory CD4+ T cells trigger regulatory T cell (Tregs) responses, which prematurely suppress the naïve CD4+ T cell response, leading to a similar OAS effect in CD4+ T cells. This mechanism is illustrated through a mathematical model incorporating naïve and memory CD4+ T cell proliferation, interleukin-2 (IL-2), and Tregs. The model, calibrated with experimental data, employs numerical simulations to analyse how CD4+ T cell responses vary with the degree of cross-reactivity between memory CD4+ T cells and the antigen associated with the secondary infection. The findings indicate that the immune response is weakest at an intermediate level of cross-reactivity, a key characteristic of OAS. This mechanism may also explain OAS in antibody responses.
Collapse
Affiliation(s)
- Mingran Zhang
- College of Information Science and Technology, Donghua University, Shanghai, China
| | - Junling Ma
- Department of Mathematics and Statistics, University of Victoria, Victoria, British Columbia, Canada
| | - Meili Li
- School of Mathematics and Statistics, Donghua University, Shanghai, China
| |
Collapse
|
2
|
Maurer DP, Vu M, Schmidt AG. Antigenic drift expands influenza viral escape pathways from recalled humoral immunity. Immunity 2025; 58:716-727.e6. [PMID: 40023162 PMCID: PMC11906258 DOI: 10.1016/j.immuni.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/16/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
Initial exposure to a rapidly evolving virus establishes B cell memory that biases later responses to antigenically drifted strains. This "immune imprinting" implies that subsequent exposure to a drifted strain can induce affinity maturation of memory B cells toward cross-reactivity with the drifted strain and hence toward greater overall breadth. Here, we used deep mutational scanning of H1 influenza hemagglutinins (HAs) to investigate how viruses evolve in response to these broad antibody response. We identified escape mutations from clonal antibody lineages that targeted the receptor binding site and lateral patch. By adjusting the antigen-antibody contacts, antibody affinity maturation restricted the potential escape routes for the eliciting strain. However, escape occurred readily in drifted strains. We attribute this escape-prone property of the drifted strains to epistatic networks within HA. Our data explain how the influenza virus continues to evolve in the human population by escaping even broad antibody responses.
Collapse
Affiliation(s)
- Daniel P Maurer
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mya Vu
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Aaron G Schmidt
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Xu Z, Peng Q, Xu J, Zhang H, Song J, Wei D, Zeng Q. Dynamic modeling of antibody repertoire reshaping in response to viral infections. Comput Biol Med 2025; 184:109475. [PMID: 39616881 DOI: 10.1016/j.compbiomed.2024.109475] [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: 06/19/2024] [Revised: 11/09/2024] [Accepted: 11/24/2024] [Indexed: 12/22/2024]
Abstract
For decades, research has largely focused on the generation of high-affinity, antigen-specific antibodies during viral infections. This emphasis has made it challenging for immunologists to systematically evaluate the mechanisms initiating humoral immunity in specific immune responses. In this study, we employ ordinary differential equations (ODE) to investigate the dynamic reshaping of the entire antibody repertoire in response to viral infections. Our findings demonstrate that the host's antibody atlas undergoes significant restructuring during these infections by the selective expansion of antibody pools with strong binding activity. The simulation results indicate that the ELISA (Enzyme-Linked Immunosorbent Assay) outcomes do not directly reflect the levels of specific neutralizing antibodies, but rather represent a quantitative response of the reshaped antibody repertoire following infection. Our model transcends traditional theories of immune memory, providing an explanation for the sustained presence of specific antibodies in the human body in long term. Additionally, our model extends to explore the mechanistic basis of the original antigenic sin, providing practical applications of our framework. One important application of this model is that it indicates that antibodies with a faster forward binding rate are more effective in preventing and treating associated viral infections compared to those with higher binding affinity.
Collapse
Affiliation(s)
- Zhaobin Xu
- Department of Life Science, Dezhou University, Dezhou 253023, China.
| | - Qingzhi Peng
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China
| | - Junxiao Xu
- Department of Life Science, Dezhou University, Dezhou 253023, China
| | - Hongmei Zhang
- Department of Life Science, Dezhou University, Dezhou 253023, China
| | - Jian Song
- Department of Life Science, Dezhou University, Dezhou 253023, China
| | - Dongqing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China; Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan, 473006, China; Peng Cheng National Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, Shenzhen, Guangdong, 518055, China
| | - Qiangcheng Zeng
- Department of Life Science, Dezhou University, Dezhou 253023, China
| |
Collapse
|
4
|
Maltseva M, Keeshan A, Cooper C, Langlois MA. Immune imprinting: The persisting influence of the first antigenic encounter with rapidly evolving viruses. Hum Vaccin Immunother 2024; 20:2384192. [PMID: 39149872 PMCID: PMC11328881 DOI: 10.1080/21645515.2024.2384192] [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: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Curtis Cooper
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
5
|
Cobey S. Vaccination against rapidly evolving pathogens and the entanglements of memory. Nat Immunol 2024; 25:2015-2023. [PMID: 39384979 DOI: 10.1038/s41590-024-01970-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/26/2024] [Indexed: 10/11/2024]
Abstract
Immune memory determines infection risk and responses to future infections and vaccinations over potentially decades of life. Despite its centrality, the dynamics of memory to antigenically variable pathogens remains poorly understood. This Review examines how past exposures shape B cell responses to vaccinations with influenza and SARS-CoV-2. An overriding feature of vaccinations with these pathogens is the recall of primary responses, often termed 'imprinting' or 'original antigenic sin'. These recalled responses can inhibit the generation of new responses unless some incompletely defined conditions are met. Depending on the context, immune memory can increase or decrease the total neutralizing antibody response to variant antigens, with apparent consequences for protection. These effects are easier to measure experimentally than epidemiologically, but there is evidence that both early and recent exposures influence vaccine effectiveness. A few immunological interactions between adaptive immune responses and antigens might explain the seemingly discrepant effects of memory. Overall, the complex observations point to a need for more quantitative approaches to integrate high-dimensional immune data from populations with diverse exposure histories. Such approaches could help identify optimal vaccination strategies against antigenically diverse pathogens.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
6
|
Greenspan NS. Jonathan Yewdell Discusses Viral Immunology, Vaccine Development, Navigating a Scientific Career, and Offers Perspectives on Transforming Scientific Publishing and Research Education. Pathog Immun 2024; 9:94-134. [PMID: 39381058 PMCID: PMC11460944 DOI: 10.20411/pai.v9i2.753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 10/10/2024] Open
Abstract
In this interview, Jonathan Yewdell talks with Pathogens and Immunity senior editor Neil Green-span about the evolution of viral immunology, highlighting his work and the contributions of other influential scientists. He emphasizes the importance of passion and collaboration in scientific research, illustrating the potential for groundbreaking discoveries through networking. He provides advice on navigating a scientific career, stressing the significance of strong mentorship. And he shares his perspective on transforming the scientific publishing industry and research education.
Collapse
|
7
|
Ding X, Zhao F, Liu Z, Yao J, Yu H, Zhang X. Original antigenic sin: A potential double-edged effect for vaccine improvement. Biomed Pharmacother 2024; 178:117187. [PMID: 39084082 DOI: 10.1016/j.biopha.2024.117187] [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: 06/07/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Original antigenic sin (OAS) influences the immune response to subsequent infections with related variants following initial pathogen exposure. This phenomenon is characterized by cross-reactivity, which, although it may worsen infections, also provides a degree of protection against immune evasion caused by variations. This paradox complicates the development of creating universal vaccinations, as they frequently show diminished effectiveness against these emerging variants. This review aims to elucidate the diverse impacts of OAS on the immune response to various infections, emphasizing the complicated balance between beneficial and harmful outcomes. Moreover, we evaluate the influence of adjuvants and other variables on the extent of OAS, hence affecting the effectiveness of vaccines. Understanding the mechanisms of OAS that cause persistent infections and evasion of the immune system is crucial for the developing innovative vaccines. And it has significant potential for clinical applications.
Collapse
Affiliation(s)
- Xuan Ding
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Feijun Zhao
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China; Laboratory Medicine Center, the First Affiliated Hospital of University of South ChinaHengyang 421001, PR China
| | - Zhaoping Liu
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Jiangchen Yao
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Han Yu
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Xiaohong Zhang
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China.
| |
Collapse
|
8
|
McGough L, Cobey S. A speed limit on serial strain replacement from original antigenic sin. Proc Natl Acad Sci U S A 2024; 121:e2400202121. [PMID: 38857397 PMCID: PMC11194583 DOI: 10.1073/pnas.2400202121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/06/2024] [Indexed: 06/12/2024] Open
Abstract
Many pathogens evolve to escape immunity, yet it remains difficult to predict whether immune pressure will lead to diversification, serial replacement of one variant by another, or more complex patterns. Pathogen strain dynamics are mediated by cross-protective immunity, whereby exposure to one strain partially protects against infection by antigenically diverged strains. There is growing evidence that this protection is influenced by early exposures, a phenomenon referred to as original antigenic sin (OAS) or imprinting. In this paper, we derive constraints on the emergence of the pattern of successive strain replacements demonstrated by influenza, SARS-CoV-2, seasonal coronaviruses, and other pathogens. We find that OAS implies that the limited diversity found with successive strain replacement can only be maintained if [Formula: see text] is less than a threshold set by the characteristic antigenic distances for cross-protection and for the creation of new immune memory. This bound implies a "speed limit" on the evolution of new strains and a minimum variance of the distribution of infecting strains in antigenic space at any time. To carry out this analysis, we develop a theoretical model of pathogen evolution in antigenic space that implements OAS by decoupling the antigenic distances required for protection from infection and strain-specific memory creation. Our results demonstrate that OAS can play an integral role in the emergence of strain structure from host immune dynamics, preventing highly transmissible pathogens from maintaining serial strain replacement without diversification.
Collapse
Affiliation(s)
- Lauren McGough
- Department of Ecology and EvolutionThe University of Chicago, Chicago, IL60637
| | - Sarah Cobey
- Department of Ecology and EvolutionThe University of Chicago, Chicago, IL60637
| |
Collapse
|
9
|
McGough L, Cobey S. A speed limit on serial strain replacement from original antigenic sin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574172. [PMID: 38260288 PMCID: PMC10802292 DOI: 10.1101/2024.01.04.574172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Many pathogens evolve to escape immunity, yet it remains difficult to predict whether immune pressure will lead to diversification, serial replacement of one variant by another, or more complex patterns. Pathogen strain dynamics are mediated by cross-protective immunity, whereby exposure to one strain partially protects against infection by antigenically diverged strains. There is growing evidence that this protection is influenced by early exposures, a phenomenon referred to as original antigenic sin (OAS) or imprinting. In this paper, we derive new constraints on the emergence of the pattern of successive strain replacements demonstrated by influenza, SARS-CoV-2, seasonal coronaviruses, and other pathogens. We find that OAS implies that the limited diversity found with successive strain replacement can only be maintained if R 0 is less than a threshold set by the characteristic antigenic distances for cross-protection and for the creation of new immune memory. This bound implies a "speed limit" on the evolution of new strains and a minimum variance of the distribution of infecting strains in antigenic space at any time. To carry out this analysis, we develop a theoretical model of pathogen evolution in antigenic space that implements OAS by decoupling the antigenic distances required for protection from infection and strain-specific memory creation. Our results demonstrate that OAS can play an integral role in the emergence of strain structure from host immune dynamics, preventing highly transmissible pathogens from maintaining serial strain replacement without diversification.
Collapse
Affiliation(s)
- Lauren McGough
- Department of Ecology and Evolution, University of Chicago, Chicago, IL
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL
| |
Collapse
|
10
|
Maurer DP, Vu M, Schmidt AG. Antigenic drift expands viral escape pathways from imprinted host humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585891. [PMID: 38562862 PMCID: PMC10983950 DOI: 10.1101/2024.03.20.585891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
An initial virus exposure can imprint antibodies such that future responses to antigenically drifted strains are dependent on the identity of the imprinting strain. Subsequent exposure to antigenically distinct strains followed by affinity maturation can guide immune responses toward generation of cross-reactive antibodies. How viruses evolve in turn to escape these imprinted broad antibody responses is unclear. Here, we used clonal antibody lineages from two human donors recognizing conserved influenza virus hemagglutinin (HA) epitopes to assess viral escape potential using deep mutational scanning. We show that even though antibody affinity maturation does restrict the number of potential escape routes in the imprinting strain through repositioning the antibody variable domains, escape is still readily observed in drifted strains and attributed to epistatic networks within HA. These data explain how influenza virus continues to evolve in the human population by escaping even broad antibody responses.
Collapse
|
11
|
Hasan J, Bok S. Plasmonic Fluorescence Sensors in Diagnosis of Infectious Diseases. BIOSENSORS 2024; 14:130. [PMID: 38534237 DOI: 10.3390/bios14030130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
The increasing demand for rapid, cost-effective, and reliable diagnostic tools in personalized and point-of-care medicine is driving scientists to enhance existing technology platforms and develop new methods for detecting and measuring clinically significant biomarkers. Humanity is confronted with growing risks from emerging and recurring infectious diseases, including the influenza virus, dengue virus (DENV), human immunodeficiency virus (HIV), Ebola virus, tuberculosis, cholera, and, most notably, SARS coronavirus-2 (SARS-CoV-2; COVID-19), among others. Timely diagnosis of infections and effective disease control have always been of paramount importance. Plasmonic-based biosensing holds the potential to address the threat posed by infectious diseases by enabling prompt disease monitoring. In recent years, numerous plasmonic platforms have risen to the challenge of offering on-site strategies to complement traditional diagnostic methods like polymerase chain reaction (PCR) and enzyme-linked immunosorbent assays (ELISA). Disease detection can be accomplished through the utilization of diverse plasmonic phenomena, such as propagating surface plasmon resonance (SPR), localized SPR (LSPR), surface-enhanced Raman scattering (SERS), surface-enhanced fluorescence (SEF), surface-enhanced infrared absorption spectroscopy, and plasmonic fluorescence sensors. This review focuses on diagnostic methods employing plasmonic fluorescence sensors, highlighting their pivotal role in swift disease detection with remarkable sensitivity. It underscores the necessity for continued research to expand the scope and capabilities of plasmonic fluorescence sensors in the field of diagnostics.
Collapse
Affiliation(s)
- Juiena Hasan
- Department of Electrical and Computer Engineering, Ritchie School of Engineering and Computer Science, University of Denver, Denver, CO 80208, USA
| | - Sangho Bok
- Department of Electrical and Computer Engineering, Ritchie School of Engineering and Computer Science, University of Denver, Denver, CO 80208, USA
| |
Collapse
|
12
|
Han AX, de Jong SPJ, Russell CA. Co-evolution of immunity and seasonal influenza viruses. Nat Rev Microbiol 2023; 21:805-817. [PMID: 37532870 DOI: 10.1038/s41579-023-00945-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 08/04/2023]
Abstract
Seasonal influenza viruses cause recurring global epidemics by continually evolving to escape host immunity. The viral constraints and host immune responses that limit and drive the evolution of these viruses are increasingly well understood. However, it remains unclear how most of these advances improve the capacity to reduce the impact of seasonal influenza viruses on human health. In this Review, we synthesize recent progress made in understanding the interplay between the evolution of immunity induced by previous infections or vaccination and the evolution of seasonal influenza viruses driven by the heterogeneous accumulation of antibody-mediated immunity in humans. We discuss the functional constraints that limit the evolution of the viruses, the within-host evolutionary processes that drive the emergence of new virus variants, as well as current and prospective options for influenza virus control, including the viral and immunological barriers that must be overcome to improve the effectiveness of vaccines and antiviral drugs.
Collapse
Affiliation(s)
- Alvin X Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Simon P J de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Colin A Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Global Health, School of Public Health, Boston University, Boston, MA, USA.
| |
Collapse
|
13
|
Kastenschmidt JM, Sureshchandra S, Jain A, Hernandez-Davies JE, de Assis R, Wagoner ZW, Sorn AM, Mitul MT, Benchorin AI, Levendosky E, Ahuja G, Zhong Q, Trask D, Boeckmann J, Nakajima R, Jasinskas A, Saligrama N, Davies DH, Wagar LE. Influenza vaccine format mediates distinct cellular and antibody responses in human immune organoids. Immunity 2023; 56:1910-1926.e7. [PMID: 37478854 PMCID: PMC10433940 DOI: 10.1016/j.immuni.2023.06.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/11/2023] [Accepted: 06/20/2023] [Indexed: 07/23/2023]
Abstract
Highly effective vaccines elicit specific, robust, and durable adaptive immune responses. To advance informed vaccine design, it is critical that we understand the cellular dynamics underlying responses to different antigen formats. Here, we sought to understand how antigen-specific B and T cells were activated and participated in adaptive immune responses within the mucosal site. Using a human tonsil organoid model, we tracked the differentiation and kinetics of the adaptive immune response to influenza vaccine and virus modalities. Each antigen format elicited distinct B and T cell responses, including differences in their magnitude, diversity, phenotype, function, and breadth. These differences culminated in substantial changes in the corresponding antibody response. A major source of antigen format-related variability was the ability to recruit naive vs. memory B and T cells to the response. These findings have important implications for vaccine design and the generation of protective immune responses in the upper respiratory tract.
Collapse
Affiliation(s)
- Jenna M Kastenschmidt
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Suhas Sureshchandra
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Aarti Jain
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Jenny E Hernandez-Davies
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Rafael de Assis
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Zachary W Wagoner
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Andrew M Sorn
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Mahina Tabassum Mitul
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Aviv I Benchorin
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Elizabeth Levendosky
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO 63112, USA
| | - Gurpreet Ahuja
- Department of Pediatric Otolaryngology, Children's Hospital of Orange County, Orange, CA 92868, USA; Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, Orange, CA 92868, USA
| | - Qiu Zhong
- Department of Pediatric Otolaryngology, Children's Hospital of Orange County, Orange, CA 92868, USA; Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, Orange, CA 92868, USA
| | - Douglas Trask
- Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, Orange, CA 92868, USA
| | - Jacob Boeckmann
- Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, Orange, CA 92868, USA
| | - Rie Nakajima
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Algimantas Jasinskas
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Naresha Saligrama
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO 63112, USA; Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO 63112, USA; Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, MO 63112, USA
| | - D Huw Davies
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA
| | - Lisa E Wagar
- Department of Physiology & Biophysics, University of California Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92617, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92617, USA; Vaccine R&D Center, University of California Irvine, Irvine, CA 92617, USA.
| |
Collapse
|
14
|
Daulagala P, Mann BR, Leung K, Lau EHY, Yung L, Lei R, Nizami SIN, Wu JT, Chiu SS, Daniels RS, Wu NC, Wentworth D, Peiris M, Yen HL. Imprinted Anti-Hemagglutinin and Anti-Neuraminidase Antibody Responses after Childhood Infections of A(H1N1) and A(H1N1)pdm09 Influenza Viruses. mBio 2023; 14:e0008423. [PMID: 37070986 PMCID: PMC10294682 DOI: 10.1128/mbio.00084-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/29/2023] [Indexed: 04/19/2023] Open
Abstract
Immune imprinting is a driver known to shape the anti-hemagglutinin (HA) antibody landscape of individuals born within the same birth cohort. With the HA and neuraminidase (NA) proteins evolving at different rates under immune selection pressures, anti-HA and anti-NA antibody responses since childhood influenza virus infections have not been evaluated in parallel at the individual level. This is partly due to the limited knowledge of changes in NA antigenicity, as seasonal influenza vaccines have focused on generating neutralizing anti-HA antibodies against HA antigenic variants. Here, we systematically characterized the NA antigenic variants of seasonal A(H1N1) viruses from 1977 to 1991 and completed the antigenic profile of N1 NAs from 1977 to 2015. We identified that NA proteins of A/USSR/90/77, A/Singapore/06/86, and A/Texas/36/91 were antigenically distinct and mapped N386K as a key determinant of the NA antigenic change from A/USSR/90/77 to A/Singapore/06/86. With comprehensive panels of HA and NA antigenic variants of A(H1N1) and A(H1N1)pdm09 viruses, we determined hemagglutinin inhibition (HI) and neuraminidase inhibition (NI) antibodies from 130 subjects born between 1950 and 2015. Age-dependent imprinting was observed for both anti-HA and anti-NA antibodies, with the peak HI and NI titers predominantly detected from subjects at 4 to 12 years old during the year of initial virus isolation, except the age-independent anti-HA antibody response against A(H1N1)pdm09 viruses. More participants possessed antibodies that reacted to multiple antigenically distinct NA proteins than those with antibodies that reacted to multiple antigenically distinct HA proteins. Our results support the need to include NA proteins in seasonal influenza vaccine preparations. IMPORTANCE Seasonal influenza vaccines have aimed to generate neutralizing anti-HA antibodies for protection since licensure. More recently, anti-NA antibodies have been established as an additional correlate of protection. While HA and NA antigenic changes occurred discordantly, the anti-HA and anti-NA antibody profiles have rarely been analyzed in parallel at the individual level, due to the limited knowledge on NA antigenic changes. By characterizing NA antigenic changes of A(H1N1) viruses, we determined the anti-HA and anti-NA antibody landscape against antigenically distinct A(H1N1) and A(H1N1)pdm09 viruses using sera of 130 subjects born between 1950 and 2015. We observed age-dependent imprinting of both anti-HA and anti-NA antibodies against strains circulated during the first decade of life. A total of 67.7% (88/130) and 90% (117/130) of participants developed cross-reactive antibodies to multiple HA and NA antigens at titers ≥1:40. With slower NA antigenic changes and cross-reactive anti-NA antibody responses, including NA protein in influenza vaccine preparation may enhance vaccine efficacy.
Collapse
Affiliation(s)
- Pavithra Daulagala
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Brian R. Mann
- WHO Collaborating Center for Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Kathy Leung
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
- University of Hong Kong, Shenzhen Hospital, Shenzhen, China
| | - Eric H. Y. Lau
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Louise Yung
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Ruipeng Lei
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sarea I. N. Nizami
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Joseph T. Wu
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Susan S. Chiu
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital and Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Rodney S. Daniels
- Francis Crick Institute, Crick Worldwide Influenza Centre, WHO Collaborating Centre for Reference and Research on Influenza, London, UK
| | - Nicholas C. Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - David Wentworth
- WHO Collaborating Center for Surveillance, Epidemiology and Control of Influenza, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection (C2I), Hong Kong Science Park, Hong Kong SAR, China
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
15
|
Xie Y, Tian X, Zhang X, Yao H, Wu N. Immune interference in effectiveness of influenza and COVID-19 vaccination. Front Immunol 2023; 14:1167214. [PMID: 37153582 PMCID: PMC10154574 DOI: 10.3389/fimmu.2023.1167214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023] Open
Abstract
Vaccines are known to function as the most effective interventional therapeutics for controlling infectious diseases, including polio, smallpox, rabies, tuberculosis, influenza and SARS-CoV-2. Smallpox has been eliminated completely and polio is almost extinct because of vaccines. Rabies vaccines and Bacille Calmette-Guérin (BCG) vaccines could effectively protect humans against respective infections. However, both influenza vaccines and COVID-19 vaccines are unable to eliminate these two infectious diseases of their highly variable antigenic sites in viral proteins. Vaccine effectiveness (VE) could be negatively influenced (i.e., interfered with) by immune imprinting of previous infections or vaccinations, and repeated vaccinations could interfere with VE against infections due to mismatch between vaccine strains and endemic viral strains. Moreover, VE could also be interfered with when more than one kind of vaccine is administrated concomitantly (i.e., co-administrated), suggesting that the VE could be modulated by the vaccine-induced immunity. In this review, we revisit the evidence that support the interfered VE result from immune imprinting or repeated vaccinations in influenza and COVID-19 vaccine, and the interference in co-administration of these two types of vaccines is also discussed. Regarding the development of next-generation COVID-19 vaccines, the researchers should focus on the induction of cross-reactive T-cell responses and naive B-cell responses to overcome negative effects from the immune system itself. The strategy of co-administrating influenza and COVID-19 vaccine needs to be considered more carefully and more clinical data is needed to verify this strategy to be safe and immunogenic.
Collapse
Affiliation(s)
- Yiwen Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Xuebin Tian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Xiaodi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Nanping Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| |
Collapse
|
16
|
Einav T, Kosikova M, Radvak P, Kuo YC, Kwon HJ, Xie H. Mapping the Antibody Repertoires in Ferrets with Repeated Influenza A/H3 Infections: Is Original Antigenic Sin Really "Sinful"? Viruses 2023; 15:374. [PMID: 36851590 PMCID: PMC9959794 DOI: 10.3390/v15020374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The influenza-specific antibody repertoire is continuously reshaped by infection and vaccination. The host immune response to contemporary viruses can be redirected to preferentially boost antibodies specific for viruses encountered early in life, a phenomenon called original antigenic sin (OAS) that is suggested to be responsible for diminished vaccine effectiveness after repeated seasonal vaccination. Using a new computational tool called Neutralization Landscapes, we tracked the progression of hemagglutination inhibition antibodies within ferret antisera elicited by repeated influenza A/H3 infections and deciphered the influence of prior exposures on the de novo antibody response to evolved viruses. The results indicate that a broadly neutralizing antibody signature can nevertheless be induced by repeated exposures despite OAS induction. Our study offers a new way to visualize how immune history shapes individual antibodies within a repertoire, which may help to inform future universal influenza vaccine design.
Collapse
Affiliation(s)
- Tal Einav
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Martina Kosikova
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Peter Radvak
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yuan-Chia Kuo
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hyung Joon Kwon
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hang Xie
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
17
|
Long Z, He J, Shuai Q, Zhang K, Xiang J, Wang H, Xie S, Wang S, Du W, Yao X, Huang J. Influenza vaccination-induced H3 stalk-reactive memory B-cell clone expansion. Vaccine 2023; 41:1132-1141. [PMID: 36621409 DOI: 10.1016/j.vaccine.2022.12.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023]
Abstract
Current vaccine formulations elicit a recall immune response against viruses by targeting epitopes on the globular head of hemagglutinin (HA), and stalk-reactive antibodies are rarely found. However, stalk-specific memory B-cell expansion after influenza vaccination is poorly understood. In this study, B cells were isolated from individuals immunized with seasonal tetravalent influenza vaccines at days 0 and 28 for H7N9 stimulation in vitro. Plasma and supernatants were collected for the analysis of anti-HA IgG using ELISA and a Luminex assay. Memory B cells were positively enriched, and total RNA was extracted for B cell receptor (BCR) H-CDR3 sequencing. All subjects displayed increased anti-H3 antibody secretion after vaccination, whereas no increase in cH5/3-reactive IgG levels was detected. The number of shared memory B-cell clones among individuals dropped dramatically from 593 to 37. Four out of 5 subjects displayed enhanced frequencies of the VH3-23 and VH3-30 genes, and one exhibited an increase in the frequency of VH1-18, which are associated with the stalk of HA. An increase in H3 stalk-specific antibodies produced by B cells stimulated with H7N9 viruses was detected after vaccination. These results demonstrated that H3 stalk-specific memory B cells can expand and secrete antibodies that bind to the stalk in vitro, although no increase in serum H3 stalk-reactive antibodies was found after vaccination, indicating potential for developing a universal vaccine strategy.
Collapse
Affiliation(s)
- Zhaoyi Long
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiang He
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Blood Transfusion, Suining Central Hospital, Suining, China
| | - Qinglu Shuai
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ke Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jim Xiang
- Cancer Research Cluster, Saskatchewan Cancer Agency, Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Huan Wang
- Key Laboratory of Infectious Disease and Biosafety, Provincial Department of Education, Guizhou, Zunyi Medical University, Zunyi, China
| | - Shuang Xie
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shengyu Wang
- Key Laboratory of Infectious Disease and Biosafety, Provincial Department of Education, Guizhou, Zunyi Medical University, Zunyi, China
| | - Wensheng Du
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xinsheng Yao
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Junqiong Huang
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
18
|
Chen Y, Hilchey SP, Wang J, Garigen J, Zand MS, Huang J. Anamnestic broadly reactive antibodies induced by H7N9 virus more efficiently bind to seasonal H3N2 strains. Hum Vaccin Immunother 2022; 18:2128014. [PMID: 36197079 DOI: 10.1080/21645515.2022.2128014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The very first influenza virus exposure in a human during infancy is known to imprint the host immune system. However, it is unclear how the memory B cells that first target virus epitopes affect antibody response to the stalk of hemagglutinin (HA) domain of influenza virus. Our study is designed to measure the cross-reactivity of antibodies induced by inactivated H7N9 virus using isolated human peripheral blood B cells. Most of the participants displayed higher levels of plasma IgG against the seasonal strains A/Vic11 and A/Cali09 than those binding to historical outbreak A/HK68 and A/PR8. H3 stalk-binding antibodies were detected in plasma at a 1:5000 dilution in 12 of 13 donors, H1 stalk-binding antibodies in all donors, indicating the existence of H3 and H1 stalk-reactive memory B cells. A moderate to high level of broadly cross-reactive antibodies was induced in memory B cells from all donors after in vitro stimulation of B cells with H7N9 virus. H3 stalk-binding antibodies were also detected in most subjects, with cross-reactivity to H1 and H7 stalk domains. The stalk-reactive antibodies bound to five H3 strains spanning 45 years and different H1, H2, H3, H5, H6, H7, H9 and B strains. Interestingly, H1- and H3-reactive IgG were much higher than H7-binding antibodies after 6 days of H7N9 stimulation. Our results demonstrate that HA stalk-reactive antibodies induced by H7N9 viruses more efficiently bound to yearly circulating both H3N2 and H1N1 strains than the boosting strain, indicating that HA stalk immunological imprint can be extended across currently circulating strains or vaccines.
Collapse
Affiliation(s)
- Yao Chen
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Shannon P Hilchey
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jiong Wang
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jessica Garigen
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, USA
| | - Martin S Zand
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, USA
| | - Junqiong Huang
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
19
|
Desikan R, Linderman SL, Davis C, Zarnitsyna VI, Ahmed H, Antia R. Vaccine models predict rules for updating vaccines against evolving pathogens such as SARS-CoV-2 and influenza in the context of pre-existing immunity. Front Immunol 2022; 13:985478. [PMID: 36263031 PMCID: PMC9574365 DOI: 10.3389/fimmu.2022.985478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Currently, vaccines for SARS-CoV-2 and influenza viruses are updated if the new vaccine induces higher antibody-titers to circulating variants than current vaccines. This approach does not account for complex dynamics of how prior immunity skews recall responses to the updated vaccine. We: (i) use computational models to mechanistically dissect how prior immunity influences recall responses; (ii) explore how this affects the rules for evaluating and deploying updated vaccines; and (iii) apply this to SARS-CoV-2. Our analysis of existing data suggests that there is a strong benefit to updating the current SARS-CoV-2 vaccines to match the currently circulating variants. We propose a general two-dose strategy for determining if vaccines need updating as well as for vaccinating high-risk individuals. Finally, we directly validate our model by reanalysis of earlier human H5N1 influenza vaccine studies.
Collapse
Affiliation(s)
- Rajat Desikan
- Clinical Pharmacology Modeling & Simulation, GlaxoSmithKline (GSK), Stevenage, Hertfordshire, United Kingdom
| | - Susanne L. Linderman
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Carl Davis
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | | | - Hasan Ahmed
- Department of Biology, Emory University, Atlanta, GA, United States
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA, United States
| |
Collapse
|
20
|
Capão A, Aguiar-Oliveira ML, Caetano BC, Neves TK, Resende PC, Almeida WAF, Miranda MD, Martins-Filho OA, Brown D, Siqueira MM, Garcia CC. Analysis of Viral and Host Factors on Immunogenicity of 2018, 2019, and 2020 Southern Hemisphere Seasonal Trivalent Inactivated Influenza Vaccine in Adults in Brazil. Viruses 2022; 14:1692. [PMID: 36016313 PMCID: PMC9413331 DOI: 10.3390/v14081692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Annual vaccination against influenza is the best tool to prevent deaths and hospitalizations. Regular updates of trivalent inactivated influenza vaccines (TIV) are necessary due to high mutation rates in influenza viruses. TIV effectiveness is affected by antigenic mismatches, age, previous immunity, and other host factors. Studying TIV effectiveness annually in different populations is critical. The serological responses to Southern-Hemisphere TIV and circulating influenza strains were evaluated in 2018−2020 among Brazilian volunteers, using hemagglutination inhibition (HI) assays. Post-vaccination titers were corrected to account for pre-vaccination titers. Our population achieved >83% post-vaccination seroprotection levels, whereas seroconversion rates ranged from 10% to 46%. TIV significantly enhanced antibody titers and seroprotection against all prior and contemporary vaccine and circulating strains tested. Strong cross-reactive responses were detected, especially between H1N1 subtypes. A/Singapore/INFIMH-16-0019/2016, included in the 2018 TIV, induced the poorest response. Significant titer and seroprotection reductions were observed 6 and 12 months after vaccination. Age had a slight effect on TIV response, whereas previous vaccination was associated with lower seroconversion rates and titers. Despite this, TIV induced high seroprotection for all strains, in all groups. Regular TIV evaluations, based on regional influenza strain circulation, should be conducted and the factors affecting response studied.
Collapse
Affiliation(s)
- Artur Capão
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Maria L. Aguiar-Oliveira
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Braulia C. Caetano
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Thayssa K. Neves
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Paola C. Resende
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Walquiria A. F. Almeida
- Secretariat of Surveillance in Health (SVS), Ministry of Health (MoH), Brasília-Federal District, Rio de Janeiro 70723-040, Brazil;
| | - Milene D. Miranda
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Olindo A.ssis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, René Rachou Institute, FIOCRUZ, Belo Horizonte 30190-002, Brazil;
| | - David Brown
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
- UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Marilda M. Siqueira
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| | - Cristiana C. Garcia
- Laboratory of Respiratory Viruses and Measles, National Influenza Center (NIC)/World Health Organization (WHO), Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (A.C.); (M.L.A.-O.); (B.C.C.); (T.K.N.); (P.C.R.); (M.D.M.); (D.B.); (M.M.S.)
| |
Collapse
|
21
|
Torresi J, Edeling MA, Nolan T, Godfrey DI. A Complementary Union of SARS-CoV2 Natural and Vaccine Induced Immune Responses. Front Immunol 2022; 13:914167. [PMID: 35911696 PMCID: PMC9326230 DOI: 10.3389/fimmu.2022.914167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/13/2022] [Indexed: 12/27/2022] Open
Abstract
Our understanding of the immune responses that follow SARS-CoV-2 infection and vaccination has progressed considerably since the COVID-19 pandemic was first declared on the 11th of March in 2020. Recovery from infection is associated with the development of protective immune responses, although over time these become less effective against new emerging SARS-CoV-2 variants. Consequently, reinfection with SARS-CoV-2 variants is not infrequent and has contributed to the ongoing pandemic. COVID-19 vaccines have had a tremendous impact on reducing infection and particularly the number of deaths associated with SARS-CoV-2 infection. However, waning of vaccine induced immunity plus the emergence of new variants has necessitated the use of boosters to maintain the benefits of vaccination in reducing COVID-19 associated deaths. Boosting is also beneficial for individuals who have recovered from COVID-19 and developed natural immunity, also enhancing responses immune responses to SARS-CoV-2 variants. This review summarizes our understanding of the immune responses that follow SARS-CoV-2 infection and vaccination, the risks of reinfection with emerging variants and the very important protective role vaccine boosting plays in both vaccinated and previously infected individuals.
Collapse
Affiliation(s)
- Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Melissa A. Edeling
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Terry Nolan
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Desikan R, Linderman SL, Davis C, Zarnitsyna V, Ahmed H, Antia R. Modeling suggests that multiple immunizations or infections will reveal the benefits of updating SARS-CoV-2 vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.05.21.492928. [PMID: 35665010 PMCID: PMC9164442 DOI: 10.1101/2022.05.21.492928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
When should vaccines to evolving pathogens such as SARS-CoV-2 be updated? Our computational models address this focusing on updating SARS-CoV-2 vaccines to the currently circulating Omicron variant. Current studies typically compare the antibody titers to the new variant following a single dose of the original-vaccine versus the updated-vaccine in previously immunized individuals. These studies find that the updated-vaccine does not induce higher titers to the vaccine-variant compared with the original-vaccine, suggesting that updating may not be needed. Our models recapitulate this observation but suggest that vaccination with the updated-vaccine generates qualitatively different humoral immunity, a small fraction of which is specific for unique epitopes to the new variant. Our simulations suggest that these new variant-specific responses could dominate following subsequent vaccination or infection with either the currently circulating or future variants. We suggest a two-dose strategy for determining if the vaccine needs updating and for vaccinating high-risk individuals.
Collapse
Affiliation(s)
- Rajat Desikan
- Clinical Pharmacology Modeling & Simulation, GlaxoSmithKline (GSK), Gunnels Wood Rd, Stevenage, Hertfordshire, SG1 2NY, United Kingdom
- These authors contributed equally
| | - Susanne L. Linderman
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Carl Davis
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Veronika Zarnitsyna
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Hasan Ahmed
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA 30322, USA
- These authors contributed equally
| |
Collapse
|
23
|
Ercanoglu MS, Gieselmann L, Dähling S, Poopalasingam N, Detmer S, Koch M, Korenkov M, Halwe S, Klüver M, Di Cristanziano V, Janicki H, Schlotz M, Worczinski J, Gathof B, Gruell H, Zehner M, Becker S, Vanshylla K, Kreer C, Klein F. No substantial preexisting B cell immunity against SARS-CoV-2 in healthy adults. iScience 2022; 25:103951. [PMID: 35224466 PMCID: PMC8857777 DOI: 10.1016/j.isci.2022.103951] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/22/2021] [Accepted: 02/16/2022] [Indexed: 11/29/2022] Open
Abstract
Preexisting immunity against SARS-CoV-2 may have critical implications for our understanding of COVID-19 susceptibility and severity. The presence and clinical relevance of a preexisting B cell immunity remain to be fully elucidated. Here, we provide a detailed analysis of the B cell immunity to SARS-CoV-2 in unexposed individuals. To this end, we extensively investigated SARS-CoV-2 humoral immunity in 150 adults sampled pre-pandemically. Comprehensive screening of donor plasma and purified IgG samples for binding and neutralization in various functional assays revealed no substantial activity against SARS-CoV-2 but broad reactivity to endemic betacoronaviruses. Moreover, we analyzed antibody sequences of 8,174 putatively SARS-CoV-2-reactive B cells at a single cell level and generated and tested 158 monoclonal antibodies. None of these antibodies displayed relevant binding or neutralizing activity against SARS-CoV-2. Taken together, our results show no evidence of competent preexisting antibody and B cell immunity against SARS-CoV-2 in unexposed adults. Comprehensive analysis of the B cell response to SARS-CoV-2 in pre-pandemic samples No substantial plasma and IgG reactivity against SARS-CoV-2 MAbs isolated from pre-pandemic samples showed no SARS-CoV-2 neutralizing activity No indication of competent preexisting B cell immunity against SARS-CoV-2
Collapse
Affiliation(s)
- Meryem Seda Ercanoglu
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Lutz Gieselmann
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Sabrina Dähling
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Nareshkumar Poopalasingam
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Susanne Detmer
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Manuel Koch
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.,Institute for Dental Research and Oral Musculoskeletal Biology and Center for Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Michael Korenkov
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Sandro Halwe
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35042 Marburg, Germany.,German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Michael Klüver
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35042 Marburg, Germany.,German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Veronica Di Cristanziano
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Hanna Janicki
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Maike Schlotz
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Johanna Worczinski
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Birgit Gathof
- Institute of Transfusion Medicine, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Henning Gruell
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Matthias Zehner
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35042 Marburg, Germany.,German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Kanika Vanshylla
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Christoph Kreer
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.,German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
24
|
Affinity maturation for an optimal balance between long-term immune coverage and short-term resource constraints. Proc Natl Acad Sci U S A 2022; 119:2113512119. [PMID: 35177475 PMCID: PMC8872716 DOI: 10.1073/pnas.2113512119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
Humoral immunity relies on the mutation and selection of B cells to better recognize pathogens. This affinity maturation process produces cells with diverse recognition capabilities. Examining optimal immune strategies that maximize the long-term immune coverage at a minimal metabolic cost, we show when the immune system should mount a de novo response rather than rely on existing memory cells. Our theory recapitulates known modes of the B cell response, predicts the empirical form of the distribution of clone sizes, and rationalizes as a trade-off between metabolic and immune costs the antigenic imprinting effects that limit the efficacy of vaccines (original antigenic sin). Our predictions provide a framework to interpret experimental results that could be used to inform vaccination strategies. In order to target threatening pathogens, the adaptive immune system performs a continuous reorganization of its lymphocyte repertoire. Following an immune challenge, the B cell repertoire can evolve cells of increased specificity for the encountered strain. This process of affinity maturation generates a memory pool whose diversity and size remain difficult to predict. We assume that the immune system follows a strategy that maximizes the long-term immune coverage and minimizes the short-term metabolic costs associated with affinity maturation. This strategy is defined as an optimal decision process on a finite dimensional phenotypic space, where a preexisting population of cells is sequentially challenged with a neutrally evolving strain. We show that the low specificity and high diversity of memory B cells—a key experimental result—can be explained as a strategy to protect against pathogens that evolve fast enough to escape highly potent but narrow memory. This plasticity of the repertoire drives the emergence of distinct regimes for the size and diversity of the memory pool, depending on the density of de novo responding cells and on the mutation rate of the strain. The model predicts power-law distributions of clonotype sizes observed in data and rationalizes antigenic imprinting as a strategy to minimize metabolic costs while keeping good immune protection against future strains.
Collapse
|
25
|
Andrews SF, Raab JE, Gorman J, Gillespie RA, Cheung CSF, Rawi R, Cominsky LY, Boyington JC, Creanga A, Shen CH, Harris DR, Olia AS, Nazzari AF, Zhou T, Houser KV, Chen GL, Mascola JR, Graham BS, Kanekiyo M, Ledgerwood JE, Kwong PD, McDermott AB. A single residue in influenza virus H2 hemagglutinin enhances the breadth of the B cell response elicited by H2 vaccination. Nat Med 2022; 28:373-382. [PMID: 35115707 DOI: 10.1038/s41591-021-01636-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/19/2021] [Indexed: 11/09/2022]
Abstract
Conserved epitopes on the influenza hemagglutinin (HA) stem are an attractive target for universal vaccine strategies as they elicit broadly neutralizing antibodies. Such antibody responses to stem-specific epitopes have been extensively characterized for HA subtypes H1 and H5 in humans. H2N2 influenza virus circulated 50 years ago and represents a pandemic threat due to the lack of widespread immunity, but, unlike H1 and H5, the H2 HA stem contains Phe45HA2 predicted to sterically clash with HA stem-binding antibodies characterized to date. To understand the effect of Phe45HA2, we compared the HA stem-specific B cell response in post hoc analyses of two phase 1 clinical trials, one testing vaccination with an H2 ferritin nanoparticle immunogen ( NCT03186781 ) and one with an inactivated H5N1 vaccine ( NCT01086657 ). In H2-naive individuals, the magnitude of the B cell response was equivalent, but H2-elicited HA stem-binding B cells displayed greater cross-reactivity than those elicited by H5. However, in individuals with childhood H2 exposure, H5-elicited HA stem-binding B cells also displayed high cross-reactivity, suggesting recall of memory B cells formed 50 years ago. Overall, we propose that a one-residue difference on an HA immunogen can alter establishment and expansion of broadly neutralizing memory B cells. These data have implications for stem-based universal influenza vaccination strategies.
Collapse
Affiliation(s)
- Sarah F Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Julie E Raab
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca A Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Crystal S F Cheung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Reda Rawi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Y Cominsky
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey C Boyington
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adrian Creanga
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chen-Hsiang Shen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Darcy R Harris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alexandra F Nazzari
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Katherine V Houser
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Grace L Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Oidtman RJ, Arevalo P, Bi Q, McGough L, Russo CJ, Vera Cruz D, Costa Vieira M, Gostic KM. Influenza immune escape under heterogeneous host immune histories. Trends Microbiol 2021; 29:1072-1082. [PMID: 34218981 PMCID: PMC8578193 DOI: 10.1016/j.tim.2021.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022]
Abstract
In a pattern called immune imprinting, individuals gain the strongest immune protection against the influenza strains encountered earliest in life. In many recent examples, differences in early infection history can explain birth year-associated differences in susceptibility (cohort effects). Susceptibility shapes strain fitness, but without a clear conceptual model linking host susceptibility to the identity and order of past infections general conclusions on the evolutionary and epidemic implications of cohort effects are not possible. Failure to differentiate between cohort effects caused by differences in the set, rather than the order (path), of past infections is a current source of confusion. We review and refine hypotheses for path-dependent cohort effects, which include imprinting. We highlight strategies to measure their underlying causes and emergent consequences.
Collapse
Affiliation(s)
- Rachel J Oidtman
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Philip Arevalo
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Qifang Bi
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Lauren McGough
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | | | - Diana Vera Cruz
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Marcos Costa Vieira
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Katelyn M Gostic
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
27
|
Guthmiller JJ, Utset HA, Wilson PC. B Cell Responses against Influenza Viruses: Short-Lived Humoral Immunity against a Life-Long Threat. Viruses 2021; 13:965. [PMID: 34067435 PMCID: PMC8224597 DOI: 10.3390/v13060965] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022] Open
Abstract
Antibodies are critical for providing protection against influenza virus infections. However, protective humoral immunity against influenza viruses is limited by the antigenic drift and shift of the major surface glycoproteins, hemagglutinin and neuraminidase. Importantly, people are exposed to influenza viruses throughout their life and tend to reuse memory B cells from prior exposure to generate antibodies against new variants. Despite this, people tend to recall memory B cells against constantly evolving variable epitopes or non-protective antigens, as opposed to recalling them against broadly neutralizing epitopes of hemagglutinin. In this review, we discuss the factors that impact the generation and recall of memory B cells against distinct viral antigens, as well as the immunological limitations preventing broadly neutralizing antibody responses. Lastly, we discuss how next-generation vaccine platforms can potentially overcome these obstacles to generate robust and long-lived protection against influenza A viruses.
Collapse
Affiliation(s)
- Jenna J. Guthmiller
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
| | - Henry A. Utset
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
| | - Patrick C. Wilson
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
28
|
Abstract
We review the phenomenon of "original antigenic sin" (OAS) in antibody responses to influenza A virus (IAV) infection or vaccination. OAS refers to the preferential induction of antibodies with higher affinity to priming versus boosting immunogens. We emphasize its mechanistic basis and origins in the basic immunobiology of B-cell responses to myriad immunogens. We tabulate 23 studies in animals and humans to show that the magnitude of OAS depends on many variables. We discuss a number of misconceptions about OAS, examine the extent to which OAS is sinful, and argue that OAS is evolutionary selected and not a deleterious by-product of selection for other features of the immune response. We end by raising questions regarding the mechanistic basis of OAS whose answers could contribute to improving influenza virus vaccines on the road to the holy grail of a "universal" influenza vaccine.
Collapse
Affiliation(s)
- Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jefferson J S Santos
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
29
|
Schnaack OH, Nourmohammad A. Optimal evolutionary decision-making to store immune memory. eLife 2021; 10:61346. [PMID: 33908347 PMCID: PMC8116052 DOI: 10.7554/elife.61346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/23/2021] [Indexed: 12/25/2022] Open
Abstract
The adaptive immune system provides a diverse set of molecules that can mount specific responses against a multitude of pathogens. Memory is a key feature of adaptive immunity, which allows organisms to respond more readily upon re-infections. However, differentiation of memory cells is still one of the least understood cell fate decisions. Here, we introduce a mathematical framework to characterize optimal strategies to store memory to maximize the utility of immune response over an organism's lifetime. We show that memory production should be actively regulated to balance between affinity and cross-reactivity of immune receptors for an effective protection against evolving pathogens. Moreover, we predict that specificity of memory should depend on the organism's lifespan, and shorter lived organisms with fewer pathogenic encounters should store more cross-reactive memory. Our framework provides a baseline to gauge the efficacy of immune memory in light of an organism's coevolutionary history with pathogens.
Collapse
Affiliation(s)
- Oskar H Schnaack
- Max Planck Institute for Dynamics and Self-organization, Göttingen, Germany.,Department of Physics, University of Washington, Seattle, United States
| | - Armita Nourmohammad
- Max Planck Institute for Dynamics and Self-organization, Göttingen, Germany.,Department of Physics, University of Washington, Seattle, United States.,Fred Hutchinson Cancer Research Center, Seattle, United States
| |
Collapse
|
30
|
Tan X, Letendre JH, Collins JJ, Wong WW. Synthetic biology in the clinic: engineering vaccines, diagnostics, and therapeutics. Cell 2021; 184:881-898. [PMID: 33571426 PMCID: PMC7897318 DOI: 10.1016/j.cell.2021.01.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/17/2022]
Abstract
Synthetic biology is a design-driven discipline centered on engineering novel biological functions through the discovery, characterization, and repurposing of molecular parts. Several synthetic biological solutions to critical biomedical problems are on the verge of widespread adoption and demonstrate the burgeoning maturation of the field. Here, we highlight applications of synthetic biology in vaccine development, molecular diagnostics, and cell-based therapeutics, emphasizing technologies approved for clinical use or in active clinical trials. We conclude by drawing attention to recent innovations in synthetic biology that are likely to have a significant impact on future applications in biomedicine.
Collapse
Affiliation(s)
- Xiao Tan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA; Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA
| | - Justin H Letendre
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA
| | - James J Collins
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Synthetic Biology Center, MIT, 77 Massachusetts Ave., Cambridge, MA 02139, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
31
|
Dugan HL, Guthmiller JJ, Arevalo P, Huang M, Chen YQ, Neu KE, Henry C, Zheng NY, Lan LYL, Tepora ME, Stovicek O, Bitar D, Palm AKE, Stamper CT, Changrob S, Utset HA, Coughlan L, Krammer F, Cobey S, Wilson PC. Preexisting immunity shapes distinct antibody landscapes after influenza virus infection and vaccination in humans. Sci Transl Med 2020; 12:eabd3601. [PMID: 33298562 PMCID: PMC8115023 DOI: 10.1126/scitranslmed.abd3601] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022]
Abstract
Humans are repeatedly exposed to variants of influenza virus throughout their lifetime. As a result, preexisting influenza-specific memory B cells can dominate the response after infection or vaccination. Memory B cells recalled by adulthood exposure are largely reactive to conserved viral epitopes present in childhood strains, posing unclear consequences on the ability of B cells to adapt to and neutralize newly emerged strains. We sought to investigate the impact of preexisting immunity on generation of protective antibody responses to conserved viral epitopes upon influenza virus infection and vaccination in humans. We accomplished this by characterizing monoclonal antibodies (mAbs) from plasmablasts, which are predominantly derived from preexisting memory B cells. We found that, whereas some influenza infection-induced mAbs bound conserved and neutralizing epitopes on the hemagglutinin (HA) stalk domain or neuraminidase, most of the mAbs elicited by infection targeted non-neutralizing epitopes on nucleoprotein and other unknown antigens. Furthermore, most infection-induced mAbs had equal or stronger affinity to childhood strains, indicating recall of memory B cells from childhood exposures. Vaccination-induced mAbs were similarly induced from past exposures and exhibited substantial breadth of viral binding, although, in contrast to infection-induced mAbs, they targeted neutralizing HA head epitopes. Last, cocktails of infection-induced mAbs displayed reduced protective ability in mice compared to vaccination-induced mAbs. These findings reveal that both preexisting immunity and exposure type shape protective antibody responses to conserved influenza virus epitopes in humans. Natural infection largely recalls cross-reactive memory B cells against non-neutralizing epitopes, whereas vaccination harnesses preexisting immunity to target protective HA epitopes.
Collapse
Affiliation(s)
- Haley L Dugan
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Jenna J Guthmiller
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Philip Arevalo
- Department of Ecology and Evolution, University of Chicago, Chicago, IL 60637, USA
| | - Min Huang
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Yao-Qing Chen
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Karlynn E Neu
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Carole Henry
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Nai-Ying Zheng
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Linda Yu-Ling Lan
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Micah E Tepora
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Olivia Stovicek
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Dalia Bitar
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Anna-Karin E Palm
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | | | - Siriruk Changrob
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Henry A Utset
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL 60637, USA
| | - Patrick C Wilson
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
32
|
The Effects of Imprinting and Repeated Seasonal Influenza Vaccination on Adaptive Immunity after Influenza Vaccination. Vaccines (Basel) 2020; 8:vaccines8040663. [PMID: 33171854 PMCID: PMC7712189 DOI: 10.3390/vaccines8040663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 11/17/2022] Open
Abstract
(1) Background: The influenza virus continues to cause significant annual morbidity and mortality. The overall efficacy of seasonal influenza vaccination is suboptimal, which is partly due to host immune factors. The effects of imprinting and repeated seasonal influenza vaccination were investigated to assess for immune factors and mechanisms that impact influenza vaccine responses. (2) Methods: Twenty participants were enrolled into a prospective pilot study based on birth cohort and seasonal influenza immunization history. Immunologic parameters were assessed over a six-month period after the seasonal influenza vaccine was administered. (3) Results: There was no significant imprinting effect, as measured by hemagglutination inhibition (HAI) fold change, HAI geometric mean titer (GMT) for Day 29 or Day 180 post-vaccination and antigen- specific antibody-secreting cells (ASC) for Day 8 post-vaccination. Individuals who had minimal prior seasonal influenza vaccination had a higher magnitude ASC response and a higher HAI fold change post-vaccination than individuals who were repeatedly vaccinated. (4) Conclusions: Repeated seasonal influenza vaccination resulted in a decreased fold change of the immune response, although individuals in this cohort tended to have high HAI titers at baseline that persisted after vaccination. Imprinting effects were not observed in this cohort. These host immune factors should be considered in the development of universal influenza vaccines. ClinicalTrials.gov Identifier: NCT03686514.
Collapse
|
33
|
Worobey M, Plotkin S, Hensley SE. Influenza Vaccines Delivered in Early Childhood Could Turn Antigenic Sin into Antigenic Blessings. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a038471. [PMID: 31964642 DOI: 10.1101/cshperspect.a038471] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Substantial evidence has accumulated that "original antigenic sin" is a central factor shaping immune responses against influenza viruses. Here, we argue that this propensity of initial influenza virus exposure to establish a lifelong immunological imprint presents a remarkable opportunity: Immunization of infants prior to their initial, natural viral exposure could circumvent narrow immunological imprinting directed toward a single viral strain. Simultaneous initial exposure to antigens from multiple influenza strains via vaccination holds the promise of extending immunological imprinting across all currently circulating strains and against potential pandemic strains of influenza A virus, potentially providing a readily accessible form of universal protection against severe disease from both pandemic and seasonal influenza.
Collapse
Affiliation(s)
- Michael Worobey
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, Arizona 85721, USA
| | | | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
34
|
Original antigenic sin priming of influenza virus hemagglutinin stalk antibodies. Proc Natl Acad Sci U S A 2020; 117:17221-17227. [PMID: 32631992 DOI: 10.1073/pnas.1920321117] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Immunity to influenza viruses can be long-lived, but reinfections with antigenically distinct viral strains and subtypes are common. Reinfections can boost antibody responses against viral strains first encountered in childhood through a process termed "original antigenic sin." It is unknown how initial childhood exposures affect the induction of antibodies against the hemagglutinin (HA) stalk domain of influenza viruses. This is an important consideration since broadly reactive HA stalk antibodies can protect against infection, and universal vaccine platforms are being developed to induce these antibodies. Here we show that experimentally infected ferrets and naturally infected humans establish strong "immunological imprints" against HA stalk antigens first encountered during primary influenza virus infections. We found that HA stalk antibodies are surprisingly boosted upon subsequent infections with antigenically distinct influenza A virus subtypes. Paradoxically, these heterosubtypic-boosted HA stalk antibodies do not bind efficiently to the boosting influenza virus strain. Our results demonstrate that an individual's HA stalk antibody response is dependent on the specific subtype of influenza virus that they first encounter early in life. We propose that humans are susceptible to heterosubtypic influenza virus infections later in life since these viruses boost HA stalk antibodies that do not bind efficiently to the boosting antigen.
Collapse
|
35
|
Park JK, Xiao Y, Ramuta MD, Rosas LA, Fong S, Matthews AM, Freeman AD, Gouzoulis MA, Batchenkova NA, Yang X, Scherler K, Qi L, Reed S, Athota R, Czajkowski L, Han A, Morens DM, Walters KA, Memoli MJ, Kash JC, Taubenberger JK. Pre-existing immunity to influenza virus hemagglutinin stalk might drive selection for antibody-escape mutant viruses in a human challenge model. Nat Med 2020; 26:1240-1246. [PMID: 32601336 DOI: 10.1038/s41591-020-0937-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 05/08/2020] [Indexed: 01/07/2023]
Abstract
The conserved region of influenza hemagglutinin (HA) stalk (or stem) has gained attention as a potent target for universal influenza vaccines1-5. Although the HA stalk region is relatively well conserved, the evolutionarily dynamic nature of influenza viruses6 raises concerns about the possible emergence of viruses carrying stalk escape mutation(s) under sufficient immune pressure. Here we show that immune pressure on the HA stalk can lead to expansion of escape mutant viruses in study participants challenged with a 2009 H1N1 pandemic influenza virus inoculum containing an A388V polymorphism in the HA stalk (45% wild type and 55% mutant). High level of stalk antibody titers was associated with the selection of the mutant virus both in humans and in vitro. Although the mutant virus showed slightly decreased replication in mice, it was not observed in cell culture, ferrets or human challenge participants. The A388V mutation conferred resistance to some of the potent HA stalk broadly neutralizing monoclonal antibodies (bNAbs). Co-culture of wild-type and mutant viruses in the presence of either a bNAb or human serum resulted in rapid expansion of the mutant. These data shed light on a potential obstacle for the success of HA-stalk-targeting universal influenza vaccines-viral escape from vaccine-induced stalk immunity.
Collapse
Affiliation(s)
- Jae-Keun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mitchell D Ramuta
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sharon Fong
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alexis M Matthews
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ashley D Freeman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Monica A Gouzoulis
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Natalia A Batchenkova
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xingdong Yang
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Li Qi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Susan Reed
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rani Athota
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Czajkowski
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alison Han
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David M Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Matthew J Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
36
|
Pardi N, Hogan MJ, Weissman D. Recent advances in mRNA vaccine technology. Curr Opin Immunol 2020; 65:14-20. [PMID: 32244193 DOI: 10.1016/j.coi.2020.01.008] [Citation(s) in RCA: 313] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
Messenger RNA (mRNA) vaccines represent a relatively new vaccine class showing great promise for the future. This optimism is built on recently published studies demonstrating the efficacy of mRNA vaccines in combatting several types of cancer and infectious pathogens where conventional vaccine platforms may fail to induce protective immune responses. These results would not have been possible without critical recent innovations in the field, such as the development of safe and efficient materials for in vivo mRNA delivery and advanced protocols for the production of high quality mRNA. This review summarizes the most important developments in mRNA vaccines from the past few years and discusses the challenges and future directions for the field.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Michael J Hogan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Lyashchenko KP, Vordermeier HM, Waters WR. Memory B cells and tuberculosis. Vet Immunol Immunopathol 2020; 221:110016. [PMID: 32050091 DOI: 10.1016/j.vetimm.2020.110016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023]
Abstract
Immunological memory is a central feature of adaptive immunity. Memory B cells are generated upon stimulation with antigen presented by follicular dendritic cells in the peripheral lymphoid tissues. This process typically involves class-switch recombination and somatic hypermutation and it can be dependent or independent on germinal centers or T cell help. The mature B cell memory pool is generally characterized by remarkable heterogeneity of functionally and phenotypically distinct sub-populations supporting multi-layer immune plasticity. Memory B cells found in human patients infected with Mycobacterium tuberculosis include IgD+ CD27+ and IgM+ CD27+ subsets. In addition, expansion of atypical memory B cells characterized by the lack of CD27 expression and by inability to respond to antigen-induced re-activation is documented in human tuberculosis. These functionally impaired memory B cells are believed to have adverse effects on host immunity. Human and animal studies demonstrate recruitment of antigen-activated B cells to the infection sites and their presence in lung granulomas where proliferating B cells are organized into discrete clusters resembling germinal centers of secondary lymphoid organs. Cattle studies show development of IgM+, IgG+, and IgA+ memory B cells in M. bovis infection with the ability to rapidly differentiate into antibody-producing plasma cells upon antigen re-exposure. This review discusses recent advances in research on generation, re-activation, heterogeneity, and immunobiological functions of memory B cells in tuberculosis. The role of memory B cells in post-skin test recall antibody responses in bovine tuberculosis and implications for development of improved immunodiagnostics are also reviewed.
Collapse
Affiliation(s)
| | - H Martin Vordermeier
- Tuberculosis Research Group, Animal and Plant Health Agency, Addlestone, United Kingdom; Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, US Department of Agriculture, Ames, IA, USA
| |
Collapse
|
38
|
Krammer F, Weir JP, Engelhardt O, Katz JM, Cox RJ. Meeting report and review: Immunological assays and correlates of protection for next-generation influenza vaccines. Influenza Other Respir Viruses 2019; 14:237-243. [PMID: 31837101 PMCID: PMC7040967 DOI: 10.1111/irv.12706] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND This report summarizes the discussions and conclusions from the "Immunological Assays and Correlates of Protection for Next-Generation Influenza Vaccines" meeting which took place in Siena, Italy, from March 31, 2019, to April 2, 2019. CONCLUSIONS Furthermore, we review current correlates of protection against influenza virus infection and disease and their usefulness for the development of next generation broadly protective and universal influenza virus vaccines.
Collapse
Affiliation(s)
- Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Jerry P Weir
- Division of Viral Products, Food and Drug Administration, Bethesda, MD, USA
| | - Othmar Engelhardt
- Division of Virology, National Institute for Biological Standards and Control, Potters Bar, UK
| | - Jacqueline M Katz
- Formerly Influenza Branch, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Rebecca Jane Cox
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| |
Collapse
|
39
|
Cobey S, Gouma S, Parkhouse K, Chambers BS, Ertl HC, Schmader KE, Halpin RA, Lin X, Stockwell TB, Das SR, Landon E, Tesic V, Youngster I, Pinsky BA, Wentworth DE, Hensley SE, Grad YH. Poor Immunogenicity, Not Vaccine Strain Egg Adaptation, May Explain the Low H3N2 Influenza Vaccine Effectiveness in 2012-2013. Clin Infect Dis 2019; 67:327-333. [PMID: 29471464 PMCID: PMC6051447 DOI: 10.1093/cid/ciy097] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 02/03/2018] [Indexed: 12/28/2022] Open
Abstract
Background Influenza vaccination aims to prevent infection by influenza virus and reduce associated morbidity and mortality; however, vaccine effectiveness (VE) can be modest, especially for subtype A(H3N2). Low VE has been attributed to mismatches between the vaccine and circulating influenza strains and to the vaccine’s elicitation of protective immunity in only a subset of the population. The low H3N2 VE in the 2012–2013 season was attributed to egg-adaptive mutations that created antigenic mismatch between the actual vaccine strain (IVR-165) and both the intended vaccine strain (A/Victoria/361/2011) and the predominant circulating strains (clades 3C.2 and 3C.3). Methods We investigated the basis of low VE in 2012–2013 by determining whether vaccinated and unvaccinated individuals were infected by different viral strains and by assessing the serologic responses to IVR-165, A/Victoria/361/2011, and 3C.2 and 3C.3 strains in an adult cohort before and after vaccination. Results We found no significant genetic differences between the strains that infected vaccinated and unvaccinated individuals. Vaccination increased titers to A/Victoria/361/2011 and 3C.2 and 3C.3 representative strains as much as to IVR-165. These results are consistent with the hypothesis that vaccination boosted cross-reactive immune responses instead of specific responses against unique vaccine epitopes. Only approximately one-third of the cohort achieved a ≥4-fold increase in titer. Conclusions In contrast to analyses based on ferret studies, low H3N2 VE in 2012–2013 in adults does not appear to be due to egg adaptation of the vaccine strain. Instead, low VE might have been caused by low vaccine immunogenicity in a subset of the population.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology & Evolution, University of Chicago, Illinois
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kaela Parkhouse
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Benjamin S Chambers
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Hildegund C Ertl
- Division of Geriatrics, Duke University Medical Center, North Carolina.,Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, North Carolina
| | - Kenneth E Schmader
- Division of Geriatrics, Duke University Medical Center, North Carolina.,Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, North Carolina
| | - Rebecca A Halpin
- Department of Infectious Disease, J. Craig Venter Institute, Rockville, Maryland
| | - Xudong Lin
- Department of Infectious Disease, J. Craig Venter Institute, Rockville, Maryland
| | - Timothy B Stockwell
- Department of Infectious Disease, J. Craig Venter Institute, Rockville, Maryland
| | - Suman R Das
- Department of Infectious Disease, J. Craig Venter Institute, Rockville, Maryland
| | - Emily Landon
- Department of Medicine, University of Chicago, Illinois
| | - Vera Tesic
- Department of Pathology, University of Chicago, Illinois
| | - Ilan Youngster
- Division of Pediatrics and the Center for Microbiome Research, Assaf Harofeh Medical Center, Tel Aviv University, Israel.,Division of Infectious Diseases, Boston Children's Hospital, Massachusetts
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University School of Medicine, California.,Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, California
| | - David E Wentworth
- Department of Infectious Disease, J. Craig Venter Institute, Rockville, Maryland
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Yonatan H Grad
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, Massachusetts.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
40
|
Skowronski DM, Sabaiduc S, Leir S, Rose C, Zou M, Murti M, Dickinson JA, Olsha R, Gubbay JB, Croxen MA, Charest H, Bastien N, Li Y, Jassem A, Krajden M, De Serres G. Paradoxical clade- and age-specific vaccine effectiveness during the 2018/19 influenza A(H3N2) epidemic in Canada: potential imprint-regulated effect of vaccine (I-REV). Euro Surveill 2019; 24:1900585. [PMID: 31771709 PMCID: PMC6864978 DOI: 10.2807/1560-7917.es.2019.24.46.1900585] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/04/2019] [Indexed: 11/20/2022] Open
Abstract
IntroductionThe Canadian Sentinel Practitioner Surveillance Network reports vaccine effectiveness (VE) for the 2018/19 influenza A(H3N2) epidemic.AimTo explain a paradoxical signal of increased clade 3C.3a risk among 35-54-year-old vaccinees, we hypothesise childhood immunological imprinting and a cohort effect following the 1968 influenza A(H3N2) pandemic.MethodsWe assessed VE by test-negative design for influenza A(H3N2) overall and for co-circulating clades 3C.2a1b and 3C.3a. VE variation by age in 2018/19 was compared with amino acid variation in the haemagglutinin glycoprotein by year since 1968.ResultsInfluenza A(H3N2) VE was 17% (95% CI: -13 to 39) overall: 27% (95% CI: -7 to 50) for 3C.2a1b and -32% (95% CI: -119 to 21) for 3C.3a. Among 20-64-year-olds, VE was -7% (95% CI: -56 to 26): 6% (95% CI: -49 to 41) for 3C.2a1b and -96% (95% CI: -277 to -2) for 3C.3a. Clade 3C.3a VE showed a pronounced negative dip among 35-54-year-olds in whom the odds of medically attended illness were > 4-fold increased for vaccinated vs unvaccinated participants (p < 0.005). This age group was primed in childhood to influenza A(H3N2) viruses that for two decades following the 1968 pandemic bore a serine at haemagglutinin position 159, in common with contemporary 3C.3a viruses but mismatched to 3C.2a vaccine strains instead bearing tyrosine.DiscussionImprinting by the first childhood influenza infection is known to confer long-lasting immunity focused toward priming epitopes. Our findings suggest vaccine mismatch may negatively interact with imprinted immunity. The immunological mechanisms for imprint-regulated effect of vaccine (I-REV) warrant investigation.
Collapse
Affiliation(s)
- Danuta M Skowronski
- British Columbia Centre for Disease Control, Vancouver, Canada
- University of British Columbia, Vancouver, Canada
| | - Suzana Sabaiduc
- British Columbia Centre for Disease Control, Vancouver, Canada
| | - Siobhan Leir
- British Columbia Centre for Disease Control, Vancouver, Canada
| | - Caren Rose
- British Columbia Centre for Disease Control, Vancouver, Canada
- University of British Columbia, Vancouver, Canada
| | - Macy Zou
- British Columbia Centre for Disease Control, Vancouver, Canada
| | - Michelle Murti
- Public Health Ontario, Toronto, Canada
- University of Toronto, Toronto, Canada
| | | | | | - Jonathan B Gubbay
- Public Health Ontario, Toronto, Canada
- University of Toronto, Toronto, Canada
| | - Matthew A Croxen
- Alberta Precision Laboratories, Edmonton, Alberta
- University of Alberta, Edmonton, Canada
| | - Hugues Charest
- Institut National de Santé Publique du Québec, Québec, Canada
| | - Nathalie Bastien
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Yan Li
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Agatha Jassem
- British Columbia Centre for Disease Control, Vancouver, Canada
- University of British Columbia, Vancouver, Canada
| | - Mel Krajden
- British Columbia Centre for Disease Control, Vancouver, Canada
- University of British Columbia, Vancouver, Canada
| | - Gaston De Serres
- Laval University, Quebec, Canada
- Centre Hospitalier Universitaire de Québec, Québec, Canada
- Institut National de Santé Publique du Québec, Québec, Canada
| |
Collapse
|
41
|
Kosikova M, Li L, Radvak P, Ye Z, Wan XF, Xie H. Imprinting of Repeated Influenza A/H3 Exposures on Antibody Quantity and Antibody Quality: Implications for Seasonal Vaccine Strain Selection and Vaccine Performance. Clin Infect Dis 2019; 67:1523-1532. [PMID: 29672713 DOI: 10.1093/cid/ciy327] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/12/2018] [Indexed: 01/04/2023] Open
Abstract
Background Reduced seasonal influenza vaccine effectiveness (VE) was observed in individuals who received repeated annual vaccinations. Preexisting influenza antibody levels were also found inversely correlated with postvaccination titers. These reports suggest that preexisting immunity may affect contemporary seasonal vaccine performance. Methods Influenza A/H3 specific cross-reactivity of postvaccination sera from humans with or without preexisting immunity was assessed by hemagglutination inhibition (HAI) assay. Ferret antisera induced by repeated H3 exposures were also subjected to HAI, antibody affinity, and antibody avidity analyses. Results Human postvaccination sera derived from subjects with or without preexisting immunity showed different cross-reactivity against H3 variant viruses. Similarly, the breadth of cross-reactive ferret antibodies induced by repeated H3 exposures was also broadened. Antigenic differences between H3 viruses characterized by ferret antisera became smaller as the number of exposures increased. Although repeated H3 exposures induced "original antigenic sin" phenomena in HAI titers against later exposed viruses, resultant ferret antibodies showed gradually enhanced avidity for different H3/hemagglutinin. Increased antibody avidity was found to be inversely correlated with decreased antigenic differences among H3 viruses characterized. Conclusions Our results suggest that repeated H3 exposures imprinted not only antibody quantity but also antibody quality. The "naive" ferret model currently used for vaccine strain selection does not recapitulate the complexity of human preexisting immunity. Vaccine strains identified hereby may not provide coverage sufficient for those who were frequently infected and/or vaccinated, leading to the reduced VE observed.
Collapse
Affiliation(s)
- Martina Kosikova
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Lei Li
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University
| | - Peter Radvak
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Zhiping Ye
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Xiu-Feng Wan
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University
| | - Hang Xie
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
42
|
Historical H1N1 Influenza Virus Imprinting Increases Vaccine Protection by Influencing the Activity and Sustained Production of Antibodies Elicited at Vaccination in Ferrets. Vaccines (Basel) 2019; 7:vaccines7040133. [PMID: 31569351 PMCID: PMC6963198 DOI: 10.3390/vaccines7040133] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022] Open
Abstract
Influenza virus imprinting is now understood to significantly influence the immune responses and clinical outcome of influenza virus infections that occur later in life. Due to the yearly cycling of influenza viruses, humans are imprinted with the circulating virus of their birth year and subsequently build a complex influenza virus immune history. Despite this knowledge, little is known about how the imprinting strain influences vaccine responses. To investigate the immune responses of the imprinted host to split-virion vaccination, we imprinted ferrets with a sublethal dose of the historical seasonal H1N1 strain A/USSR/90/1977. After a +60-day recovery period to build immune memory, ferrets were immunized and then challenged on Day 123. Antibody specificity and recall were investigated throughout the time course. At challenge, the imprinted vaccinated ferrets did not experience significant disease, while naïve-vaccinated ferrets had significant weight loss. Haemagglutination inhibition assays showed that imprinted ferrets had a more robust antibody response post vaccination and increased virus neutralization activity. Imprinted-vaccinated animals had increased virus-specific IgG antibodies compared to the other experimental groups, suggesting B-cell maturity and plasticity at vaccination. These results should be considered when designing the next generation of influenza vaccines.
Collapse
|
43
|
Padilla-Quirarte HO, Lopez-Guerrero DV, Gutierrez-Xicotencatl L, Esquivel-Guadarrama F. Protective Antibodies Against Influenza Proteins. Front Immunol 2019; 10:1677. [PMID: 31379866 PMCID: PMC6657620 DOI: 10.3389/fimmu.2019.01677] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022] Open
Abstract
The influenza A virus infection continues to be a threat to the human population. The seasonal variation of the virus and the likelihood of periodical pandemics caused by completely new virus strains make it difficult to produce vaccines that efficiently protect against this infection. Antibodies (Abs) are very important in preventing the infection and in blocking virus propagation once the infection has taken place. However, the precise protection mechanism provided by these Abs still needs to be established. Furthermore, most research has focused on Abs directed to the globular head domain of hemagglutinin (HA). However, other domains of HA (like the stem) and other proteins are also able to elicit protective Ab responses. In this article, we review the current knowledge about the role of both neutralizing and non-neutralizing anti-influenza proteins Abs that play a protective role during infection or vaccination.
Collapse
Affiliation(s)
- Herbey O Padilla-Quirarte
- LIV, Facultad de Medicina, Universidad Autonoma del Estado de Morelos, Cuernavaca, Mexico.,Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Cuernavaca, Mexico
| | | | | | | |
Collapse
|
44
|
Auladell M, Jia X, Hensen L, Chua B, Fox A, Nguyen THO, Doherty PC, Kedzierska K. Recalling the Future: Immunological Memory Toward Unpredictable Influenza Viruses. Front Immunol 2019; 10:1400. [PMID: 31312199 PMCID: PMC6614380 DOI: 10.3389/fimmu.2019.01400] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/03/2019] [Indexed: 01/09/2023] Open
Abstract
Persistent and durable immunological memory forms the basis of any successful vaccination protocol. Generation of pre-existing memory B cell and T cell pools is thus the key for maintaining protective immunity to seasonal, pandemic and avian influenza viruses. Long-lived antibody secreting cells (ASCs) are responsible for maintaining antibody levels in peripheral blood. Generated with CD4+ T help after naïve B cell precursors encounter their cognate antigen, the linked processes of differentiation (including Ig class switching) and proliferation also give rise to memory B cells, which then can change rapidly to ASC status after subsequent influenza encounters. Given that influenza viruses evolve rapidly as a consequence of antibody-driven mutational change (antigenic drift), the current influenza vaccines need to be reformulated frequently and annual vaccination is recommended. Without that process of regular renewal, they provide little protection against “drifted” (particularly H3N2) variants and are mainly ineffective when a novel pandemic (2009 A/H1N1 “swine” flu) strain suddenly emerges. Such limitation of antibody-mediated protection might be circumvented, at least in part, by adding a novel vaccine component that promotes cross-reactive CD8+ T cells specific for conserved viral peptides, presented by widely distributed HLA types. Such “memory” cytotoxic T lymphocytes (CTLs) can rapidly be recalled to CTL effector status. Here, we review how B cells and follicular T cells are elicited following influenza vaccination and how they survive into a long-term memory. We describe how CD8+ CTL memory is established following influenza virus infection, and how a robust CTL recall response can lead to more rapid virus elimination by destroying virus-infected cells, and recovery. Exploiting long-term, cross-reactive CTL against the continuously evolving and unpredictable influenza viruses provides a possible mechanism for preventing a disastrous pandemic comparable to the 1918-1919 H1N1 “Spanish flu,” which killed more than 50 million people worldwide.
Collapse
Affiliation(s)
- Maria Auladell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Brendon Chua
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Development of Influenza B Universal Vaccine Candidates Using the "Mosaic" Hemagglutinin Approach. J Virol 2019; 93:JVI.00333-19. [PMID: 30944178 DOI: 10.1128/jvi.00333-19] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/26/2019] [Indexed: 12/19/2022] Open
Abstract
Influenza B viruses cause seasonal epidemics and are a considerable burden to public health. However, protection by current seasonal vaccines is suboptimal due to the antigenic changes of the circulating strains. In this study, we report a novel universal influenza B virus vaccination strategy based on "mosaic" hemagglutinins. We generated mosaic B hemagglutinins by replacing the major antigenic sites of the type B hemagglutinin with corresponding sequences from exotic influenza A hemagglutinins and expressed them as soluble trimeric proteins. Sequential vaccination with recombinant mosaic B hemagglutinin proteins conferred cross-protection against both homologous and heterologous influenza B virus strains in the mouse model. Of note, we rescued recombinant influenza B viruses expressing mosaic B hemagglutinins, which could serve as the basis for a universal influenza B virus vaccine.IMPORTANCE This work reports a universal influenza B virus vaccination strategy based on focusing antibody responses to conserved head and stalk epitopes of the hemagglutinin. Recombinant mosaic influenza B hemagglutinin proteins and recombinant viruses have been generated as novel vaccine candidates. This vaccine strategy provided broad cross-protection in the mouse model. Our findings will inform and drive development toward a more effective influenza B virus vaccine.
Collapse
|
46
|
Nienen M, Stervbo U, Mölder F, Kaliszczyk S, Kuchenbecker L, Gayova L, Schweiger B, Jürchott K, Hecht J, Neumann AU, Rahmann S, Westhoff T, Reinke P, Thiel A, Babel N. The Role of Pre-existing Cross-Reactive Central Memory CD4 T-Cells in Vaccination With Previously Unseen Influenza Strains. Front Immunol 2019; 10:593. [PMID: 31019503 PMCID: PMC6458262 DOI: 10.3389/fimmu.2019.00593] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Influenza vaccination is a common approach to prevent seasonal and pandemic influenza. Pre-existing antibodies against close viral strains might impair antibody formation against previously unseen strains-a process called original antigenic sin. The role of this pre-existing cellular immunity in this process is, despite some hints from animal models, not clear. Here, we analyzed cellular and humoral immunity in healthy individuals before and after vaccination with seasonal influenza vaccine. Based on influenza-specific hemagglutination inhibiting (HI) titers, vaccinees were grouped into HI-negative and -positive cohorts followed by in-depth cytometric and TCR repertoire analysis. Both serological groups revealed cross-reactive T-cell memory to the vaccine strains at baseline that gave rise to the majority of vaccine-specific T-cells post vaccination. On the contrary, very limited number of vaccine-specific T-cell clones was recruited from the naive pool. Furthermore, baseline quantity of vaccine-specific central memory helper T-cells and clonotype richness of this population directly correlated with the vaccination efficacy. Our findings suggest that the deliberate recruitment of pre-existing cross-reactive cellular memory might help to improve vaccination outcome.
Collapse
Affiliation(s)
- Mikalai Nienen
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine, Immunology and Transplantation, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Felix Mölder
- Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sviatlana Kaliszczyk
- Center for Translational Medicine, Immunology and Transplantation, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | | | | | | | - Karsten Jürchott
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Jochen Hecht
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Avidan U Neumann
- Institute of Environmental Medicine, German Research Center for Environmental Health, Helmholtz Zentrum München, Augsburg, Germany
| | - Sven Rahmann
- Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Timm Westhoff
- Department of Internal Medicine, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| | - Andreas Thiel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Center for Translational Medicine, Immunology and Transplantation, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany.,Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
47
|
Lee J, Paparoditis P, Horton AP, Frühwirth A, McDaniel JR, Jung J, Boutz DR, Hussein DA, Tanno Y, Pappas L, Ippolito GC, Corti D, Lanzavecchia A, Georgiou G. Persistent Antibody Clonotypes Dominate the Serum Response to Influenza over Multiple Years and Repeated Vaccinations. Cell Host Microbe 2019; 25:367-376.e5. [PMID: 30795981 DOI: 10.1016/j.chom.2019.01.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/02/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022]
Abstract
Humans are repeatedly exposed to influenza virus via infections and vaccinations. Understanding how multiple exposures and pre-existing immunity impact antibody responses is essential for vaccine development. Given the recent prevalence of influenza H1N1 A/California/7/2009 (CA09), we examined the clonal composition and dynamics of CA09 hemagglutinin (HA)-reactive IgG repertoire over 5 years in a donor with multiple influenza exposures. The anti-CA09 HA polyclonal response in this donor comprised 24 persistent antibody clonotypes, accounting for 72.6% ± 10.0% of the anti-CA09 HA repertoire over 5 years. These persistent antibodies displayed higher somatic hypermutation relative to transient serum antibodies detected at one time point. Additionally, persistent antibodies predominantly demonstrated cross-reactivity and potent neutralization toward a phylogenetically distant H5N1 A/Vietnam/1203/2004 (VT04) strain, a feature correlated with HA stem recognition. This analysis reveals how "serological imprinting" impacts responses to influenza and suggests that once elicited, cross-reactive antibodies targeting the HA stem can persist for years.
Collapse
Affiliation(s)
- Jiwon Lee
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Philipp Paparoditis
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland; Institute of Microbiology, ETH Zürich, Zürich 8093, Switzerland
| | - Andrew P Horton
- Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Alexander Frühwirth
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Jonathan R McDaniel
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiwon Jung
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Daniel R Boutz
- Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Dania A Hussein
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yuri Tanno
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Leontios Pappas
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Gregory C Ippolito
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | | | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland; Institute of Microbiology, ETH Zürich, Zürich 8093, Switzerland
| | - George Georgiou
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Influenza remains a major cause of morbidity and mortality. The 2017-2018 season was one of the most severe in the past decade. The exact factors determining the severity of a particular influenza season are complex and often poorly understood. RECENT FINDINGS Factors impacting annual influenza severity include characteristics of the specific virus, influenza vaccination, and antiviral use. Although viral virulence factors are important in this context and our knowledge of these is growing, there is a complex interplay between expression of these factors and their impact on a particular patient population. Vaccination has demonstrated efficacy in preventing disease, but vaccination rates remain sub-optimal and vaccine effectiveness can vary significantly between influenza strains and patient populations. Finally, while antiviral treatment is available and has shown benefits, many patients with influenza do not receive treatment. SUMMARY Strides have been made in recent years towards understanding the many factors that contribute to the severity of any particular influenza season. Obvious areas for improvement include improved vaccination rates and antiviral use. Additionally, a more complete understanding of reasons for poor strain and population-specific vaccine effectiveness may help reduce the severity of future influenza seasons.
Collapse
|
49
|
Francis ME, King ML, Kelvin AA. Back to the Future for Influenza Preimmunity-Looking Back at Influenza Virus History to Infer the Outcome of Future Infections. Viruses 2019; 11:v11020122. [PMID: 30704019 PMCID: PMC6410066 DOI: 10.3390/v11020122] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
The influenza virus-host interaction is a classic arms race. The recurrent and evolving nature of the influenza virus family allows a single host to be infected several times. Locked in co-evolution, recurrent influenza virus infection elicits continual refinement of the host immune system. Here we give historical context of circulating influenza viruses to understand how the individual immune history is mirrored by the history of influenza virus circulation. Original Antigenic Sin was first proposed as the negative influence of the host’s first influenza virus infection on the next and Imprinting modernizes Antigenic Sin incorporating both positive and negative outcomes. Building on imprinting, we refer to preimmunity as the continual refinement of the host immune system with each influenza virus infection. We discuss imprinting and the interplay of influenza virus homology, vaccination, and host age establishing preimmunity. We outline host signatures and outcomes of tandem infection according to the sequence of virus and classify these relationships as monosubtypic homologous, monosubtypic heterologous, heterosubtypic, or heterotypic sequential infections. Finally, the preimmunity knowledge gaps are highlighted for future investigation. Understanding the effects of antigenic variable recurrent influenza virus infection on immune refinement will advance vaccination strategies, as well as pandemic preparedness.
Collapse
Affiliation(s)
- Magen Ellen Francis
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada.
| | - Morgan Leslie King
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada.
| | - Alyson Ann Kelvin
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada.
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada.
- Canadian Centre for Vaccinology, IWK Health Centre, Halifax NS B3K 6R8, Canada.
| |
Collapse
|
50
|
Zhang A, Stacey HD, Mullarkey CE, Miller MS. Original Antigenic Sin: How First Exposure Shapes Lifelong Anti–Influenza Virus Immune Responses. THE JOURNAL OF IMMUNOLOGY 2019; 202:335-340. [DOI: 10.4049/jimmunol.1801149] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022]
|