1
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Blazevic A, Edwards RL, Xia M, Eickhoff CS, Hamzabegovic F, Meza KA, Ning H, Tennant J, Mosby KJ, Ritchie JC, Girmay T, Lai L, McCullough M, Beck A, Kelley C, Edupuganti S, Kabbani S, Buchanan W, Makhene MK, Voronca D, Cherikh S, Goll JB, Rouphael NG, Mulligan MJ, Hoft DF. Phase 1 Open-Label Dose Escalation Trial for the Development of a Human Bacillus Calmette-Guérin Challenge Model for Assessment of Tuberculosis Immunity In Vivo. J Infect Dis 2024; 229:1498-1508. [PMID: 38019956 PMCID: PMC11095547 DOI: 10.1093/infdis/jiad441] [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: 05/12/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND A controlled human infection model for assessing tuberculosis (TB) immunity can accelerate new vaccine development. METHODS In this phase 1 dose escalation trial, 92 healthy adults received a single intradermal injection of 2 × 106 to 16 × 106 colony-forming units of Bacillus Calmette-Guérin (BCG). The primary endpoints were safety and BCG shedding as measured by quantitative polymerase chain reaction, colony-forming unit plating, and MGIT BACTEC culture. RESULTS Doses up to 8 × 106 were safe, and there was evidence for increased BCG shedding with dose escalation. The MGIT time-to-positivity assay was the most consistent and precise measure of shedding. Power analyses indicated that 10% differences in MGIT time to positivity (area under the curve) could be detected in small cohorts (n = 30). Potential biomarkers of mycobacterial immunity were identified that correlated with shedding. Transcriptomic analysis uncovered dose- and time-dependent effects of BCG challenge and identified a putative transcriptional TB protective signature. Furthermore, we identified immunologic and transcriptomal differences that could represent an immune component underlying the observed higher rate of TB disease incidence in males. CONCLUSIONS The safety, reactogenicity, and immunogenicity profiles indicate that this BCG human challenge model is feasible for assessing in vivo TB immunity and could facilitate the vaccine development process. CLINICAL TRIALS REGISTRATION NCT01868464 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Azra Blazevic
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - Rachel L Edwards
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - Mei Xia
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | | | - Fahreta Hamzabegovic
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - Krystal A Meza
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - Huan Ning
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - Janice Tennant
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - Karla J Mosby
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| | - James C Ritchie
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Tigisty Girmay
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Lilin Lai
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Michele McCullough
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Allison Beck
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Colleen Kelley
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Srilatha Edupuganti
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Sarah Kabbani
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Wendy Buchanan
- Division of Microbiology, Immunology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Mamodikoe K Makhene
- Division of Microbiology, Immunology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Delia Voronca
- The Emmes Company, LLC, Global Head Biomedical Data Science and Bioinformatics, Rockville, Maryland
| | - Sami Cherikh
- The Emmes Company, LLC, Global Head Biomedical Data Science and Bioinformatics, Rockville, Maryland
| | - Johannes B Goll
- The Emmes Company, LLC, Global Head Biomedical Data Science and Bioinformatics, Rockville, Maryland
| | - Nadine G Rouphael
- Hope Clinic, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | | | - Daniel F Hoft
- Department of Internal Medicine, School of Medicine, Saint Louis University, Missouri
| |
Collapse
|
3
|
Oxford JS, Catchpole A, Mann A, Bell A, Noulin N, Gill D, Oxford JR, Gilbert A, Balasingam S. A Brief History of Human Challenge Studies (1900-2021) Emphasising the Virology, Regulatory and Ethical Requirements, Raison D'etre, Ethnography, Selection of Volunteers and Unit Design. Curr Top Microbiol Immunol 2024; 445:1-32. [PMID: 35704095 DOI: 10.1007/82_2022_253] [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] [Indexed: 10/18/2022]
Abstract
Venetian quarantine 400 years ago was an important public health measure. Since 1900 this has been refined to include "challenge" or deliberate infection with pathogens be they viruses, bacteria, or parasites. Our focus is virology and ranges from the early experiments in Cuba with Yellow Fever Virus to the most widespread pathogen of our current times, COVID-19. The latter has so far caused over four million deaths worldwide and 190 million cases of the disease. Quarantine and challenge were also used to investigate the Spanish Influenza of 1918 which caused over 100 million deaths. We consider here the merits of the approach, that is the speeding up of knowledge in a practical sense leading to the more rapid licensing of vaccines and antimicrobials. At the core of quarantine and challenge initiatives is the design of the unit to allow safe confinement of the pathogen and protection of the staff. Most important though is the safety of volunteers. We can see now, as in 1900, that members of our society are prepared and willing to engage in these experiments for the public good. Our ethnology study, where the investigator observed the experiment from within the quarantine, gave us the first indication of changing attitudes amongst volunteers whilst in quarantine. These quarantine experiments, referred to as challenge studies, human infection studies, or "controlled human infection models" involve thousands of clinical samples taken over two to three weeks and can provide a wealth of immunological and molecular data on the infection itself and could allow the discovery of new targets for vaccines and therapeutics. The Yellow Fever studies from 121 years ago gave the impetus for development of a successful vaccine still used today whilst also uncovering the nature of the Yellow Fever agent, namely that it was a virus. We outline how carefully these experiments are approached and the necessity to have high quality units with self-contained air-flow along with extensive personal protective equipment for nursing and medical staff. Most important is the employment of highly trained scientific, medical and nursing staff. We face a future of emerging pathogens driven by the increasing global population, deforestation, climate change, antibiotic resistance and increased global travel. These emerging pathogens may be pathogens we currently are not aware of or have not caused outbreaks historically but could also be mutated forms of known pathogens including viruses such as influenza (H7N9, H5N1 etc.) and coronaviruses. This calls for challenge studies to be part of future pandemic preparedness as an additional tool to assist with the rapid development of broad-spectrum antimicrobials, immunomodulators and new vaccines.
Collapse
Affiliation(s)
- J S Oxford
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | | | | | | | | | - D Gill
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | - J R Oxford
- Inveresk Medical Practice, Edinburgh, E21 7BP, UK
| | | | | |
Collapse
|
4
|
Rothman RE, Niforatos JD, Youbi M, Polydefkis N, Hergenroeder A, Ako MC, Lobner K, Shaw-Saliba K, Hsieh YH. A cross-sectional study of participant recruitment rates in published phase III influenza therapeutic randomized controlled trials conducted in the clinical setting. Am J Emerg Med 2022; 61:184-191. [PMID: 36174486 DOI: 10.1016/j.ajem.2022.09.003] [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: 05/11/2022] [Revised: 08/20/2022] [Accepted: 09/03/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE A recent academic-government partnership demonstrated the feasibility of utilizing Emergency Departments (ED) as a primary site for subject enrollment in clinical trials and achieved high rates of recruitment in two U.S. EDs. Given the ongoing need to test new therapeutics for influenza and other emerging infections, we sought to describe the historical rates of participant recruitment into influenza Phase III therapeutic RCTs in various clinical venues, including EDs. STUDY DESIGN A cross-sectional study was performed of influenza therapeutic Phase III RCTs published in PubMed, Embase, Scopus, and Clinicaltrials.gov from January 2000 to June 2019. MAIN OUTCOME To estimate the weighted-average number of influenza-positive participants enrolled per site per season in influenza therapeutic RCT conducted in clinical settings, and to describe basic trial site characteristics. RESULTS 47 (0.7%) of 7008 articles were included for review of which 43 of 47 (91%) included information regarding enrollment sites; of these, 2 (5%) recruited exclusively from EDs with the remainder recruiting from mixed clinical settings (inpatient, outpatient, and ED). The median enrollment per study was 326 (IQR: 110, 502.5) with a median of 11 sites per study (IQR: 2, 59.5). Included studies reported a median of 201 (IQR: 74, 344.5) confirmed influenza-positive participants per study. The pooled number of participants enrolled per site per season was 11 (95% CI: 10, 12). The pooled enrollment numbers per clinical site after excluding the two 'ED only recruitment' studies were less [10.7 (95% CI: 9.9, 11.6)] than the pooled enrollment numbers per clinical site for the two 'ED only recruitment' studies [89.5 (95% CI 89.2-89.27)]. CONCLUSION AND RELEVANCE Published RCTs evaluating influenza therapeutics in clinical settings recruit participants from multiple sites but enroll relatively few participants, per site, per season. The few ED-based studies reported recruited more subjects per site per season. Untapped opportunities likely exist for EDs to participate and/or lead therapeutic RCTs for influenza or other emerging respiratory pathogens.
Collapse
Affiliation(s)
- Richard E Rothman
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Joshua D Niforatos
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Mehdi Youbi
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Nicholas Polydefkis
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Alaina Hergenroeder
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Michele-Corinne Ako
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Katie Lobner
- Welch Medical Library, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kathryn Shaw-Saliba
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yu-Hsiang Hsieh
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America.
| |
Collapse
|
5
|
Balasingam S, Meillon S, Chui C, Mann A, La C, Weller CL, King DF, Smith E. Human infection studies: Key considerations for challenge agent development and production. Wellcome Open Res 2022; 7:140. [PMID: 35505774 PMCID: PMC9034172 DOI: 10.12688/wellcomeopenres.17869.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 12/03/2022] Open
Abstract
Human infection (or challenge) studies involve the intentional administration of a pathogen (challenge agent) to volunteers. The selection, isolation, development and production of the challenge agent is one of the first steps in developing a challenge study and critical for minimising the risk to volunteers. Regulatory oversight for this production differs globally. Manufacturing agents within a Good Manufacturing Practice (GMP) facility reduces the risk of the manufacturing process by including processes such as confirming the identity of the challenge agent and ascertaining that it's pure and free from impurities. However, in some cases it's not possible or feasible to manufacture to GMP standards, for example where the challenge agent requires an intermediate vector for growth. There is lack of clear guidance on what the minimum requirements for high-quality safe manufacture outside of GMP facilities should be and here we describe the development of a considerations document for the selection and production of challenge agents to meet this need.
Collapse
Affiliation(s)
| | - Sarah Meillon
- Infectious Diseases
- Prevention, Wellcome Trust, UK, London, N1 2BE, UK
| | | | - Alex Mann
- Global Health Innovative Technology Fund, Tokyo 106-0032, Japan
| | - Carine La
- Global Health Innovative Technology Fund, Tokyo 106-0032, Japan
| | | | - Deborah F. King
- Infectious Diseases
- Prevention, Wellcome Trust, UK, London, N1 2BE, UK
| | - Emma Smith
- HIC-Vac, Imperial College London, National Heart & Lung Institute (NHLI), London, W2 1PG, UK
| |
Collapse
|
6
|
McIlwain DR, Chen H, Rahil Z, Bidoki NH, Jiang S, Bjornson Z, Kolhatkar NS, Martinez CJ, Gaudillière B, Hedou J, Mukherjee N, Schürch CM, Trejo A, Affrime M, Bock B, Kim K, Liebowitz D, Aghaeepour N, Tucker SN, Nolan GP. Human influenza virus challenge identifies cellular correlates of protection for oral vaccination. Cell Host Microbe 2021; 29:1828-1837.e5. [PMID: 34784508 PMCID: PMC8665113 DOI: 10.1016/j.chom.2021.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/16/2021] [Accepted: 10/21/2021] [Indexed: 01/23/2023]
Abstract
Developing new influenza vaccines with improved performance and easier administration routes hinges on defining correlates of protection. Vaccine-elicited cellular correlates of protection for influenza in humans have not yet been demonstrated. A phase-2 double-blind randomized placebo and active (inactivated influenza vaccine) controlled study provides evidence that a human-adenovirus-5-based oral influenza vaccine tablet (VXA-A1.1) can protect from H1N1 virus challenge in humans. Mass cytometry characterization of vaccine-elicited cellular immune responses identified shared and vaccine-type-specific responses across B and T cells. For VXA-A1.1, the abundance of hemagglutinin-specific plasmablasts and plasmablasts positive for integrin α4β7, phosphorylated STAT5, or lacking expression of CD62L at day 8 were significantly correlated with protection from developing viral shedding following virus challenge at day 90 and contributed to an effective machine learning model of protection. These findings reveal the characteristics of vaccine-elicited cellular correlates of protection for an oral influenza vaccine.
Collapse
Affiliation(s)
- David R McIlwain
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; WCCT Global, Cypress, CA, USA.
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zainab Rahil
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Neda Hajiakhoond Bidoki
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sizun Jiang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zach Bjornson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Julien Hedou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nilanjan Mukherjee
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Schürch
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Angelica Trejo
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Kenneth Kim
- Ark Clinical Research, LLC, Long Beach, CA, USA
| | | | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
7
|
Vaz M, Timms O, Johnson AR, S RK, Ramanathan M, Vaz M. Public perceptions on Controlled Human Infection Model (CHIM) studies-a qualitative pilot study from South India. Monash Bioeth Rev 2021; 39:68-93. [PMID: 33085005 PMCID: PMC7576547 DOI: 10.1007/s40592-020-00121-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2020] [Indexed: 04/12/2023]
Abstract
Research using Controlled Human Infection Models is yet to be attempted in India. This study was conducted to understand the perceptions of the lay public and key opinion makers prior to the possible introduction of such studies in the country. 110 respondents from urban and rural Bangalore district were interviewed using qualitative research methods of Focus Group Discussions and In-depth Interviews. The data was analyzed using grounded theory. Safety was a key concern of the lay public, expressed in terms of fear of death. The notion of infecting a healthy volunteer, the possibility of continued effects beyond the study duration and the likelihood of vulnerable populations volunteering solely for monetary benefit, were ethical concerns. Public good outcomes such as effective treatments, targeted vaccines and prevention of diseases was necessary justification for such studies. However, the comprehension of this benefit was not clear among non-medical, non-technical respondents and suggestions to seek alternatives to CHIMs repeatedly arose. There was a great deal of deflection-with each constituency feeling that people other than themselves may be ideally suited as participants. Risk takers, those without dependents, the more health and research literate, financially sound and those with an altruistic bent of mind emerged as possible CHIM volunteers. While widespread awareness and advocacy about CHIM is essential, listening to plural voices is the first step in public engagement in ethically contentious areas. Continued engagement and inclusive deliberative processes are required to redeem the mistrust of the public in research and rebuild faith in regulatory systems.
Collapse
Affiliation(s)
- Manjulika Vaz
- Division of Health and Humanities, St John's Research Institute, St John's Medical College, Bangalore, 560034, India.
| | - Olinda Timms
- Division of Health and Humanities, St John's Research Institute, St John's Medical College, Bangalore, 560034, India
| | - Avita Rose Johnson
- Department of Community Health, St John's Medical College, Bangalore, India
| | - Rathna Kumari S
- Department of Community Health, St John's Medical College, Bangalore, India
| | - Mala Ramanathan
- Achutha Menon Centre for Health Science Studies, Sree Chitra Tirunal Institute for Medical Sciences and Technology Medical College, Thiruvananthapuram, Kerala, India
| | - Mario Vaz
- Division of Health and Humanities, St John's Research Institute, St John's Medical College, Bangalore, 560034, India
- Department of Physiology, St John's Medical College, Bangalore, India
| |
Collapse
|
8
|
Hooker KL, Ganusov VV. Impact of Oseltamivir Treatment on Influenza A and B Virus Dynamics in Human Volunteers. Front Microbiol 2021; 12:631211. [PMID: 33732224 PMCID: PMC7957053 DOI: 10.3389/fmicb.2021.631211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
Influenza viruses infect millions of humans every year causing an estimated 400,000 deaths globally. Due to continuous virus evolution current vaccines provide only limited protection against the flu. Several antiviral drugs are available to treat influenza infection, and one of the most commonly used drugs is oseltamivir (Tamiflu). While the mechanism of action of oseltamivir as a neuraminidase inhibitor is well-understood, the impact of oseltamivir on influenza virus dynamics in humans has been controversial. Many clinical trials with oseltamivir have been done by pharmaceutical companies such as Roche but the results of these trials until recently have been provided as summary reports or papers. Typically, such reports included median virus shedding curves for placebo and drug-treated influenza virus infected volunteers often indicating high efficacy of the early treatment. However, median shedding curves may be not accurately representing drug impact in individual volunteers. Importantly, due to public pressure clinical trials data testing oseltamivir efficacy has been recently released in the form of redacted PDF documents. We digitized and re-analyzed experimental data on influenza virus shedding in human volunteers from three previously published trials: on influenza A (1 trial) or B viruses (2 trials). Given that not all volunteers exposed to influenza viruses actually start virus shedding we found that impact of oseltamivir on the virus shedding dynamics was dependent on (i) selection of volunteers that were infected with the virus, and (ii) the detection limit in the measurement assay; both of these details were not well-articulated in the published studies. By assuming that any non-zero viral measurement is above the limit of detection we could match previously published data on median influenza A virus (flu A study) shedding but not on influenza B virus shedding (flu B study B) in human volunteers. Additional analyses confirmed that oseltamivir had an impact on the duration of shedding and overall shedding (defined as area under the curve) but this result varied by the trial. Interestingly, treatment had no impact on the rates at which shedding increased or declined with time in individual volunteers. Additional analyses showed that oseltamivir impacted the kinetics of the end of viral shedding, and in about 20-40% of volunteers that shed the virus treatment had no impact on viral shedding duration. Our results suggest an unusual impact of oseltamivir on influenza viruses shedding kinetics and caution about the use of published median data or data from a few individuals for inferences. Furthermore, we call for the need to publish raw data from critical clinical trials that can be independently analyzed.
Collapse
Affiliation(s)
- Kyla L. Hooker
- Genome Science and Technology, University of Tennessee, Knoxville, TN, United States
| | - Vitaly V. Ganusov
- Genome Science and Technology, University of Tennessee, Knoxville, TN, United States
- Department of Microbiology, University of Tennessee, Knoxville, TN, United States
- Department of Mathematics, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
9
|
McIlwain DR, Chen H, Apkarian M, Affrime M, Bock B, Kim K, Mukherjee N, Nolan GP, McNeal MM. Performance of BioFire array or QuickVue influenza A + B test versus a validation qPCR assay for detection of influenza A during a volunteer A/California/2009/H1N1 challenge study. Virol J 2021; 18:45. [PMID: 33632249 PMCID: PMC7905982 DOI: 10.1186/s12985-021-01516-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/18/2021] [Indexed: 01/01/2023] Open
Abstract
Background Influenza places a significant burden on global health and economics. Individual case management and public health efforts to mitigate the spread of influenza are both strongly impacted by our ability to accurately and efficiently detect influenza viruses in clinical samples. Therefore, it is important to understand the performance characteristics of available assays to detect influenza in a variety of settings. We provide the first report of relative performance between two products marketed to streamline detection of influenza virus in the context of a highly controlled volunteer influenza challenge study. Methods Nasopharyngeal swab samples were collected during a controlled A/California/2009/H1N1 influenza challenge study and analyzed for detection of virus shedding using a validated qRT-PCR (qPCR) assay, a sample-to-answer qRT-PCR device (BioMerieux BioFire FilmArray RP), and an immunoassay based rapid test kit (Quidel QuickVue Influenza A + B Test). Results Relative to qPCR, the sensitivity and specificity of the BioFire assay was 72.1% [63.7–79.5%, 95% confidence interval (CI)] and 93.5% (89.3–96.4%, 95% CI) respectively. For the QuickVue rapid test the sensitivity was 8.5% (4.8–13.7%, 95% CI) and specificity was 99.2% (95.6–100%, 95% CI). Conclusion Relative to qPCR, the BioFire assay had superior performance compared to rapid test in the context of a controlled influenza challenge study. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-021-01516-0.
Collapse
Affiliation(s)
- David R McIlwain
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA. .,WCCT Global, Cypress, CA, USA.
| | - Han Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | - Nilanjan Mukherjee
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Monica M McNeal
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
10
|
Rahil Z, Leylek R, Schürch CM, Chen H, Bjornson-Hooper Z, Christensen SR, Gherardini PF, Bhate SS, Spitzer MH, Fragiadakis GK, Mukherjee N, Kim N, Jiang S, Yo J, Gaudilliere B, Affrime M, Bock B, Hensley SE, Idoyaga J, Aghaeepour N, Kim K, Nolan GP, McIlwain DR. Landscape of coordinated immune responses to H1N1 challenge in humans. J Clin Invest 2020; 130:5800-5816. [PMID: 33044226 PMCID: PMC7598057 DOI: 10.1172/jci137265] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
Influenza is a significant cause of morbidity and mortality worldwide. Here we show changes in the abundance and activation states of more than 50 immune cell subsets in 35 individuals over 11 time points during human A/California/2009 (H1N1) virus challenge monitored using mass cytometry along with other clinical assessments. Peak change in monocyte, B cell, and T cell subset frequencies coincided with peak virus shedding, followed by marked activation of T and NK cells. Results led to the identification of CD38 as a critical regulator of plasmacytoid dendritic cell function in response to influenza virus. Machine learning using study-derived clinical parameters and single-cell data effectively classified and predicted susceptibility to infection. The coordinated immune cell dynamics defined in this study provide a framework for identifying novel correlates of protection in the evaluation of future influenza therapeutics.
Collapse
Affiliation(s)
- Zainab Rahil
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Rebecca Leylek
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Christian M. Schürch
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Han Chen
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Zach Bjornson-Hooper
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Shannon R. Christensen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Salil S. Bhate
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | | | - Gabriela K. Fragiadakis
- UCSF Data Science CoLab and UCSF Department of Medicine, UCSF, San Francisco, California, USA
| | - Nilanjan Mukherjee
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Nelson Kim
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Sizun Jiang
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Jennifer Yo
- ARK Clinical Research, Long Beach, California, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | | | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Kenneth Kim
- ARK Clinical Research, Long Beach, California, USA
| | - Garry P. Nolan
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - David R. McIlwain
- Department of Pathology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- WCCT Global, Cypress, California, USA
| |
Collapse
|
11
|
Convening on the influenza human viral challenge model for universal influenza vaccines, Part 2: Methodologic considerations. Vaccine 2020; 37:4830-4834. [PMID: 31362820 DOI: 10.1016/j.vaccine.2019.06.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 11/23/2022]
Abstract
In response to global interest in the development of a universal influenza vaccine, the Bill & Melinda Gates Foundation, PATH, and the Global Funders Consortium for Universal Influenza Vaccine Development convened a meeting of experts (London, UK, May 2018) to assess the role of a standardized controlled human influenza virus infection model (CHIVIM) towards the development of novel influenza vaccine candidates. This report (in two parts) summarizes those discussions and offers consensus recommendations. Part 1 covers challenge virus selection, regulatory and ethical considerations, and issues concerning standardization, access, and capacity. This article (Part 2) summarizes the discussion and recommendations concerning CHIVIM methods. The panelists identified an overall need for increased standardization of CHIVIM trials, in order to produce comparable results that can support universal vaccine licensure. Areas of discussion included study participant selection and screening, route of exposure and dose, devices for administering challenge, rescue therapy, protection of participants and institutions, clinical outcome measures, and other considerations. The panelists agreed upon specific recommendations to improve the standardization and usefulness of the model for vaccine development. Experts agreed that a research network of institutions working with a standardized CHIVIM could contribute important data to support more rapid development and licensure of novel vaccines capable of providing long-lasting protection against seasonal and pandemic influenza strains.
Collapse
|
12
|
Meeting report: Convening on the influenza human viral challenge model for universal influenza vaccines, Part 1: Value; challenge virus selection; regulatory, industry and ethical considerations; increasing standardization, access and capacity. Vaccine 2020; 37:4823-4829. [PMID: 31362819 PMCID: PMC6677912 DOI: 10.1016/j.vaccine.2019.06.080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 11/23/2022]
Abstract
In response to global interest in the development of a universal influenza vaccine, the Bill & Melinda Gates Foundation, PATH, and the Global Funders Consortium for Universal Influenza Vaccine Development convened a meeting of experts (London, UK, May 2018) to assess the role of a standardized controlled human influenza virus infection model (CHIVIM) towards the development of novel influenza vaccine candidates. This report (in two parts) summarizes those discussions and offers consensus recommendations. This article (Part 1) covers challenge virus selection, regulatory and ethical considerations, and issues concerning standardization, access, and capacity. Part 2 covers specific methodologic considerations. Current methods for influenza vaccine development and licensure require large costly field trials. The CHIVIM requires fewer subjects and the controlled setting allows for better understanding of influenza transmission and host immunogenicity. The CHIVIM can be used to identify immune predictors of disease for at-risk populations and to measure efficacy of potential vaccines for further development. Limitations to the CHIVIM include lack of standardization, limited access to challenge viruses and assays, lack of consensus regarding role of the CHIVIM in vaccine development pathway, and concerns regarding risk to study participants and community. To address these issues, the panel of experts recommended that WHO and other key stakeholders provide guidance on standardization, challenge virus selection, and risk management. A common repository of well-characterized challenge viruses, harmonized protocols, and standardized assays should be made available to researchers. A network of research institutions performing CHIVIM trials should be created, and more study sites are needed to increase capacity. Experts agreed that a research network of institutions working with a standardized CHIVIM could contribute important data to support more rapid development and licensure of novel vaccines capable of providing long-lasting protection against seasonal and pandemic influenza strains.
Collapse
|
13
|
Schaefer GO, Tam CC, Savulescu J, Voo TC. COVID-19 vaccine development: Time to consider SARS-CoV-2 challenge studies? Vaccine 2020; 38:5085-5088. [PMID: 32540271 PMCID: PMC7269942 DOI: 10.1016/j.vaccine.2020.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 01/18/2023]
Abstract
While a human challenge study holds the prospect of accelerating the development of a vaccine for the coronavirus SARS-CoV-2, it may be opposed due to risks of harm to participants and researchers. Given the increasing number of human deaths and severe disruption to lives worldwide, we argue that a SARS-CoV-2 challenge study is ethically justifiable as its social value substantially outweighs the risks. Such a study should therefore be seriously considered as part of the global research response towards the COVID-19 pandemic. In this paper, we contribute to the debate by addressing the misperception that a challenge study for the coronavirus would lower scientific and ethical standards for vaccine research and development, and examine how it could be ethically conducted. We also set out information that needs to be disclosed to prospective participants to obtain their consent.
Collapse
Affiliation(s)
- G Owen Schaefer
- Centre for Biomedical Ethics, National University of Singapore, Yong Loo Lin School of Medicine, Block MD11, Clinical Research Centre, #02-03, 10 Medical Drive, Singapore 117597, Singapore
| | - Clarence C Tam
- Saw Swee Hock School of Public Health, National University of Singapore, Tahir Foundation Building (MD1), 12 Science Drive 2, Singapore 117549, Singapore
| | - Julian Savulescu
- Oxford Uehiro Centre for Practical Ethics, University of Oxford, Murdoch Children's Research Institute, Littlegate House, St Ebbes St, Oxford OX1 1PT, UK
| | - Teck Chuan Voo
- Centre for Biomedical Ethics, National University of Singapore, Yong Loo Lin School of Medicine, Block MD11, Clinical Research Centre, #02-03, 10 Medical Drive, Singapore 117597, Singapore.
| |
Collapse
|
14
|
Quirouette C, Younis NP, Reddy MB, Beauchemin CAA. A mathematical model describing the localization and spread of influenza A virus infection within the human respiratory tract. PLoS Comput Biol 2020; 16:e1007705. [PMID: 32282797 PMCID: PMC7179943 DOI: 10.1371/journal.pcbi.1007705] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 04/23/2020] [Accepted: 01/31/2020] [Indexed: 12/20/2022] Open
Abstract
Within the human respiratory tract (HRT), virus diffuses through the periciliary fluid (PCF) bathing the epithelium. But virus also undergoes advection: as the mucus layer sitting atop the PCF is pushed along by the ciliated cell's beating cilia, the PCF and its virus content are also pushed along, upwards towards the nose and mouth. While many mathematical models (MMs) have described the course of influenza A virus (IAV) infections in vivo, none have considered the impact of both diffusion and advection on the kinetics and localization of the infection. The MM herein represents the HRT as a one-dimensional track extending from the nose down towards the lower HRT, wherein stationary cells interact with IAV which moves within (diffusion) and along with (advection) the PCF. Diffusion was found to be negligible in the presence of advection which effectively sweeps away IAV, preventing infection from disseminating below the depth at which virus first deposits. Higher virus production rates (10-fold) are required at higher advection speeds (40 μm/s) to maintain equivalent infection severity and timing. Because virus is entrained upwards, upper parts of the HRT see more virus than lower parts. As such, infection peaks and resolves faster in the upper than in the lower HRT, making it appear as though infection progresses from the upper towards the lower HRT, as reported in mice. When the spatial MM is expanded to include cellular regeneration and an immune response, it reproduces tissue damage levels reported in patients. It also captures the kinetics of seasonal and avian IAV infections, via parameter changes consistent with reported differences between these strains, enabling comparison of their treatment with antivirals. This new MM offers a convenient and unique platform from which to study the localization and spread of respiratory viral infections within the HRT.
Collapse
Affiliation(s)
| | - Nada P. Younis
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
| | - Micaela B. Reddy
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Catherine A. A. Beauchemin
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
- Interdisciplinary Theoretical and Mathematical Sciences (iTHEMS), RIKEN, Wako, Japan
- * E-mail:
| |
Collapse
|
15
|
Sloan SE, Szretter KJ, Sundaresh B, Narayan KM, Smith PF, Skurnik D, Bedard S, Trevejo JM, Oldach D, Shriver Z. Clinical and virological responses to a broad-spectrum human monoclonal antibody in an influenza virus challenge study. Antiviral Res 2020; 184:104763. [PMID: 32151645 DOI: 10.1016/j.antiviral.2020.104763] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/06/2020] [Accepted: 03/04/2020] [Indexed: 01/03/2023]
Abstract
Influenza A infections cause significant seasonal morbidity and mortality as well as periodic pandemic infections. Currently, no approved therapies exist for patients hospitalized with influenza. The efficacy of VIS410, a broadly neutralizing human immunoglobulin IgG1 monoclonal antibody engineered to bind to the stem region of group 1 and 2 influenza A hemagglutinins, was explored in experimental human influenza infection. Healthy volunteers were inoculated with influenza A/California/07/2009 (H1N1) and received a single dose of VIS410 or placebo 24 h later. Subjects were monitored for symptoms, viral shedding, and safety, including cytokine measurements. The primary efficacy endpoint was the area under the curve (AUC) of viral load (VL) in the VIS410 group versus placebo. VIS410 treatment was associated with a 76% reduction in median VL AUC as measured by qRT-PCR (p = 0.024). Similar VIS410 antiviral activity was observed by virus culture, with a 91% reduction in median VL AUC by TCID50 (p = 0.019) compared to placebo-treated volunteers. Influenza symptoms were generally mild or moderate, with a trend toward faster resolution in VIS410-treated subjects. Treatment with VIS410 was generally safe, with an increase in gastrointestinal events that were largely mitigated by pre-treatment with oral diphenhydramine (50 mg) in combination with 600 mg of ibuprofen. Transient elevation of specific cytokines (IL-8 and TNFα) were associated with gastrointestinal adverse events. Treatment with VIS410 did not interfere with the endogenous immune response to influenza A. These data indicate that VIS410 may provide therapeutic benefit in influenza A infection. TRIAL REGISTRATION: ClinicaTtrials.gov Identification NCT02468115; https://clinicaltrials.gov/ct2/show/NCT02468115?term=NCT02468115&rank=1).
Collapse
Affiliation(s)
| | - Kristy J Szretter
- Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts 02139, USA
| | | | | | | | - David Skurnik
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Institut Necker-Enfants Malades, INSERM U1151, CNRS UMR, 8253, Paris, France; Université Paris Descartes, Paris, France; Service de Microbiologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | | | | | | |
Collapse
|
16
|
Langenberg MCC, Dekkers OM, Roestenberg M. Are placebo controls necessary in controlled human infection trials for vaccines? THE LANCET. INFECTIOUS DISEASES 2020; 20:e69-e74. [PMID: 32142640 DOI: 10.1016/s1473-3099(20)30020-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/02/2020] [Accepted: 01/12/2020] [Indexed: 12/18/2022]
Abstract
Controlled human infection trials, whereby a small group of healthy participants is deliberately exposed to a pathogen under controlled circumstances, can provide preliminary data for vaccine efficacy and for the selection of the most promising candidate vaccines for field trials. Because of the potential harm to participants through the deliberate exposure to a pathogen, the use of smaller groups minimises the cumulative risk. As such, a control group that receives a placebo vaccine followed by controlled exposure to a pathogen should be scientifically well justified. As these types of trials are designed to generate consistent infection rates and thus comparable outcomes across populations and trial sites, data from past studies (historical data) could be used as a valid alternative to placebo groups. In this Personal View, we review this option and highlight the considerations for choosing historical data as a suitable control. For the widespread application of this method, responsibility for the centralisation and sharing of data from controlled human infection trials lies with the scientific community.
Collapse
Affiliation(s)
| | - Olaf M Dekkers
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands; Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
17
|
A controlled human Schistosoma mansoni infection model to advance novel drugs, vaccines and diagnostics. Nat Med 2020; 26:326-332. [PMID: 32066978 DOI: 10.1038/s41591-020-0759-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/09/2020] [Indexed: 11/08/2022]
Abstract
Schistosomiasis treatment relies on the use of a single drug, praziquantel, which is insufficient to control transmission in highly endemic areas1. Novel medicines and vaccines are urgently needed2,3. An experimental human model for schistosomiasis could accelerate the development of these products. We performed a dose-escalating clinical safety trial in 17 volunteers with male Schistosoma mansoni cercariae, which do not produce eggs (clinicaltrials.gov NCT02755324), at the Leiden University Medical Center, the Netherlands. The primary endpoints were adverse events and infectivity. We found a dose-related increase in adverse events related to acute schistosomiasis syndrome, which occurred in 9 of 17 volunteers. Overall, 5 volunteers (all 3 of the high dose group and 2 of 11 of the medium dose group) reported severe adverse events. Worm-derived circulating anodic antigen, the biomarker of the primary infection endpoint, peaked in 82% of volunteers at 3-10 weeks following exposure. All volunteers showed IgM and IgG1 seroconversion and worm-specific cytokine production by CD4+ T cells. All volunteers were cured with praziquantel provided at 12 weeks after exposure. Infection with 20 Schistosoma mansoni cercariae led to severe adverse events in 18% of volunteers and high infection rates. This infection model paves the way for fast-track product development for treatment and prevention of schistosomiasis.
Collapse
|
18
|
McShane H. Insights and challenges in tuberculosis vaccine development. THE LANCET. RESPIRATORY MEDICINE 2019; 7:810-819. [PMID: 31416767 DOI: 10.1016/s2213-2600(19)30274-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 06/30/2019] [Accepted: 07/15/2019] [Indexed: 02/09/2023]
Abstract
Tuberculosis kills more people than any other pathogen and the need for a universally effective vaccine has never been greater. An effective vaccine will be a key tool in achieving the targets set by WHO in the End TB Strategy. Tuberculosis vaccine development is difficult and slow. Substantial progress has been made in research and development of tuberculosis vaccines in the past 20 years, and two clinical trial results from 2018 provide reason for optimism. However, many challenges to the successful licensure and deployment of an effective tuberculosis vaccine remain. The development of new tools for vaccine evaluation might facilitate these processes, and continued collaborative working and sustained funding will be essential.
Collapse
Affiliation(s)
- Helen McShane
- The Jenner Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Christensen SR, Toulmin SA, Griesman T, Lamerato LE, Petrie JG, Martin ET, Monto AS, Hensley SE. Assessing the Protective Potential of H1N1 Influenza Virus Hemagglutinin Head and Stalk Antibodies in Humans. J Virol 2019; 93:e02134-18. [PMID: 30700610 PMCID: PMC6450120 DOI: 10.1128/jvi.02134-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/23/2019] [Indexed: 11/20/2022] Open
Abstract
Seasonal influenza viruses are a major cause of human disease worldwide. Most neutralizing antibodies (Abs) elicited by influenza viruses target the head domain of the hemagglutinin (HA) protein. Anti-HA head Abs can be highly potent, but they have limited breadth since the HA head is variable. There is great interest in developing new universal immunization strategies that elicit broadly neutralizing Abs against conserved regions of HA, such as the stalk domain. Although HA stalk Abs can provide protection in animal models, it is unknown if they are present at sufficient levels in humans to provide protection against naturally acquired influenza virus infections. Here, we quantified H1N1 HA head- and stalk-specific Abs in 179 adults hospitalized during the 2015-2016 influenza virus season. We found that HA head Abs, as measured by hemagglutinin inhibition (HAI) assays, were associated with protection against naturally acquired H1N1 infection. HA stalk-specific serum total IgG titers were also associated with protection, but this association was attenuated and not statistically significant after adjustment for HA head-specific Ab titers. We found slightly higher titers of HA stalk-specific IgG1 and IgA Abs in sera from uninfected participants than in sera from infected participants; however, we found no difference in serum in vitro antibody-dependent cellular cytotoxicity activity. In passive transfer experiments, sera from participants with high HAI activity efficiently protected mice, while sera with low HAI activity protected mice to a lower extent. Our data suggest that HA head Abs are more efficient at protecting against H1N1 infection than HA stalk Abs.IMPORTANCE Abs targeting the HA head of influenza viruses are often associated with protection from influenza virus infections. These Abs typically have limited breadth, since mutations frequently arise in HA head epitopes. New vaccines targeting the more conserved HA stalk domain are being developed. Abs that target the HA stalk are protective in animal models, but it is unknown if these Abs exist at protective levels in humans. Here, we completed experiments to determine if Abs against the HA head and stalk were associated with protection from naturally acquired human influenza virus infections during the 2015-2016 influenza season.
Collapse
Affiliation(s)
- Shannon R Christensen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sushila A Toulmin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Trevor Griesman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lois E Lamerato
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Joshua G Petrie
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Emily T Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Arnold S Monto
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Pelfrene E, Mura M, Cavaleiro Sanches A, Cavaleri M. Monoclonal antibodies as anti-infective products: a promising future? Clin Microbiol Infect 2019; 25:60-64. [PMID: 29715552 PMCID: PMC7128139 DOI: 10.1016/j.cmi.2018.04.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND The paucity of licensed monoclonal antibodies (mAbs) in the infectious diseases arena strongly contrasts with the ready availability of these therapeutics for use in other conditions. AIMS This narrative review aims to assess the potential of monoclonal antibody-based interventions for infectious diseases. SOURCES A review of the literature via the Medline database was performed and complemented by published official documents on licensed anti-infective mAbs. In addition, ongoing trials were identified through a search of the clinical trial registration platform ClinicalTrials.gov. CONTENT We identified the few infections for which mAbs have been added to the therapeutic armamentarium and stressed their potential in representing a readily available protection tool against biothreats and newly emerging and reemerging infectious agents. In reviewing the historical context and main features of mAbs, we assert a potentially wider applicability and cite relevant examples of ongoing therapeutic developments. Factors hindering successful introduction of mAbs on a larger scale are outlined and thoughts are offered on how to possibly address some of these limitations. IMPLICATIONS mAbs may represent important tools in treating or preventing infections occurring with reasonably sufficient prevalence to justify demand and for which existing alternatives are not deemed fully adequate. Future initiatives need to address the prohibitive costs encountered in the development process. The feasibility of more large-scale administration of alternative modalities merits further exploration. In order to ensure optimal prospect of regulatory success, an early dialogue with competent authorities is encouraged.
Collapse
Affiliation(s)
- E Pelfrene
- Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK.
| | - M Mura
- Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK
| | - A Cavaleiro Sanches
- Quality Office, Human Medicines Research & Development Support Division, European Medicines Agency, London, UK
| | - M Cavaleri
- Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK
| |
Collapse
|
21
|
Evaluation of MEDI8852, an Anti-Influenza A Monoclonal Antibody, in Treating Acute Uncomplicated Influenza. Antimicrob Agents Chemother 2018; 62:AAC.00694-18. [PMID: 30150460 DOI: 10.1128/aac.00694-18] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/17/2018] [Indexed: 12/28/2022] Open
Abstract
We evaluated MEDI8852, a human IgG1 monoclonal antibody that binds a highly conserved influenza A hemagglutinin stalk epitope, in outpatients with uncomplicated influenza A infection. A total of 126 subjects aged 18 to 65 years were enrolled during the 2015 to 2016 Northern and 2016 Southern Hemisphere seasons. Subjects with symptom onset ≤5 days before dosing were randomized to four cohorts: 750 mg (cohort 1) or 3,000 mg (cohort 2) MEDI8852 (single intravenous infusion) plus 75 mg oseltamivir, placebo plus 75 mg oseltamivir (cohort 3), and 3,000 mg MEDI8852 alone (cohort 4). Subjects were monitored through day 10 for solicited influenza symptoms, day 28 for adverse events (AEs), and day 101 for serious AEs and AEs of special interest. Nasopharyngeal samples were collected through day 7 for confirmation of influenza A infection, viral shedding, and oseltamivir and MEDI8852 susceptibility. Slightly more AEs were reported in subjects receiving MEDI8852 (cohorts 1, 2, and 4 combined: 39/93, 41.9%) than oseltamivir only (cohort 3: 10/32, 31.3%). Most AEs were mild or moderate. The most common AE was bronchitis (11/93, 11.8%; 1/32, 3.1%). The median (range) decrease in viral shedding (log10 virus genome copies/ml) was similar between the two groups (-3.58 [-6.2. 0.5]; -3.43 [-5.9, 0.9]). Genotypic analyses found a limited number of hemagglutinin and neuraminidase amino acid changes between viruses isolated before and after therapy; however, none appeared within a known oseltamivir-resistant site or MEDI8852-binding region. The safety profile of MEDI8852 supports its continued development for treatment of patients hospitalized with influenza A infection. (This study has been registered at ClinicalTrials.gov under identifier NCT02603952.).
Collapse
|
22
|
Yogaratnam J, Rito J, Kakuda TN, Fennema H, Gupta K, Jekle CA, Mitchell T, Boyce M, Sahgal O, Balaratnam G, Chanda S, Van Remoortere P, Symons JA, Fry J. Antiviral Activity, Safety, and Pharmacokinetics of AL-794, a Novel Oral Influenza Endonuclease Inhibitor: Results of an Influenza Human Challenge Study. J Infect Dis 2018; 219:177-185. [DOI: 10.1093/infdis/jiy410] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/06/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jeysen Yogaratnam
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | - Jennifer Rito
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | - Thomas N Kakuda
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | - Hein Fennema
- Janssen Research and Development, Beerse, Belgium
| | - Kusum Gupta
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | - C Andreas Jekle
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | - Toni Mitchell
- Hammersmith Medicines Research, London, United Kingdom
| | - Malcolm Boyce
- Hammersmith Medicines Research, London, United Kingdom
| | - Omair Sahgal
- Hammersmith Medicines Research, London, United Kingdom
| | - Ganesh Balaratnam
- hVIVO Services, Queen Mary BioEnterprises Innovation Centre, London, United Kingdom
| | - Sushmita Chanda
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | | | - Julian A Symons
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| | - John Fry
- Alios Biopharma, part of Janssen Pharmaceutical Companies, South San Francisco, California
| |
Collapse
|
23
|
Roestenberg M, Hoogerwerf MA, Ferreira DM, Mordmüller B, Yazdanbakhsh M. Experimental infection of human volunteers. THE LANCET. INFECTIOUS DISEASES 2018; 18:e312-e322. [PMID: 29891332 DOI: 10.1016/s1473-3099(18)30177-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 02/08/2018] [Accepted: 03/01/2018] [Indexed: 12/13/2022]
Abstract
Controlled human infection (CHI) trials, in which healthy volunteers are experimentally infected, can accelerate the development of novel drugs and vaccines for infectious diseases of global importance. The use of CHI models is expanding from around 60 studies in the 1970s to more than 120 publications in this decade, primarily for influenza, rhinovirus, and malaria. CHI trials have provided landmark data for several registered drugs and vaccines, and have generated unprecedented scientific insights. Because of their invasive nature, CHI studies demand critical ethical review according to established frameworks. CHI-associated serious adverse events are rarely reported. Novel CHI models need standardised safety data from comparable CHI models to facilitate evidence-based risk assessments, as well as funds to produce challenge inoculum according to regulatory requirements. Advances such as the principle of controlled colonisation, the expansion of models to endemic areas, and the use of genetically attenuated strains will further broaden the scope of CHI trials.
Collapse
Affiliation(s)
| | | | | | - Benjamin Mordmüller
- Institute of Tropical Medicine and German Center for Infection Research, partner site Tübingen, University of Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
24
|
Ufer M, Shakeri-Nejad K, Gardin A, Su Z, Paule I, Marbury TC, Legangneux E. Impact of siponimod on vaccination response in a randomized, placebo-controlled study. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 4:e398. [PMID: 28955715 PMCID: PMC5608565 DOI: 10.1212/nxi.0000000000000398] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 06/23/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To evaluate effects of siponimod on response to T-cell-dependent (influenza) and T-cell-independent (pneumococcal polysaccharide vaccine [PPV-23]) vaccinations in healthy participants. METHODS In this double-blind, placebo-controlled, parallel-group study, each participant underwent a 7-week treatment period and received intramuscular injections of influenza and PPV-23 vaccines (day 21). Participants were randomized to 4 treatment groups (N = 30 each) and received placebo or siponimod 2 mg once daily in concomitant, interrupted, or preceding fashion. Individual response to vaccination was defined by a ≥4-fold (influenza) antibody titer increase and by a ≥2-fold increase in serotype-specific immunoglobulin (Ig) G concentrations (PPV-23) on day 28 vs baseline. Responder rates were compared using noninferiority analysis. RESULTS Mean influenza titers were similar to placebo in the preceding and interrupted groups but lower in the concomitant group. The proportion of participants with influenza titers ≥40 four weeks after vaccination (seroprotection) was similar to placebo across all groups and antigens. In each treatment group, response criteria were met for 3 of 4 antigens including H1N1 and H3N2. A noninferior response was determined in the context of preceding treatment but not interrupted or concomitant treatment. Regarding PPV-23, approximately 90%-100% of participants exhibited a ≥2-fold increase in IgG concentrations vs baseline. Noninferior responder rates were determined for each siponimod treatment group. CONCLUSIONS Siponimod treatment had no relevant effect on antibody response to PPV-23. European Medicines Agency response criteria were essentially met for influenza, but titers were lower on concomitant treatment. Overall, these data suggest that siponimod has limited effect on the efficacy of vaccinations with neoantigens. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that in healthy persons, siponimod had limited effect on the immune response following influenza or pneumococcal vaccinations.
Collapse
Affiliation(s)
- Mike Ufer
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| | - Kasra Shakeri-Nejad
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| | - Anne Gardin
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| | - Zhenzhong Su
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| | - Ines Paule
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| | - Thomas C Marbury
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| | - Eric Legangneux
- Novartis Pharma AG (M.U., K.S.-N., A.G., Z.S., I.P., E.L.), Basel, Switzerland; and Orlando Clinical Research Center (T.C.M.), Orlando, FL
| |
Collapse
|
25
|
Corti D, Cameroni E, Guarino B, Kallewaard NL, Zhu Q, Lanzavecchia A. Tackling influenza with broadly neutralizing antibodies. Curr Opin Virol 2017; 24:60-69. [PMID: 28527859 PMCID: PMC7102826 DOI: 10.1016/j.coviro.2017.03.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/02/2017] [Accepted: 03/09/2017] [Indexed: 11/28/2022]
Abstract
Monoclonal antibodies have revolutionized the treatment of several human diseases, including cancer, autoimmunity and inflammatory conditions and represent a new frontier for the treatment of infectious diseases. In the last decade, new methods have allowed the efficient interrogation of the human antibody repertoire from influenza immune individuals and the isolation of several monoclonal antibodies capable of dealing with the high variability of influenza viruses. Here, we will provide a comprehensive overview of the specificity, antiviral and immunological mechanisms of action and development into the clinic of broadly reactive monoclonal antibodies against influenza A and B viruses.
Collapse
Affiliation(s)
| | | | | | | | - Qing Zhu
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| |
Collapse
|
26
|
Sparrow E, Friede M, Sheikh M, Torvaldsen S, Newall AT. Passive immunization for influenza through antibody therapies, a review of the pipeline, challenges and potential applications. Vaccine 2016; 34:5442-5448. [PMID: 27622299 PMCID: PMC5357764 DOI: 10.1016/j.vaccine.2016.08.057] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/09/2016] [Accepted: 08/18/2016] [Indexed: 11/24/2022]
Abstract
The Global Action Plan for influenza vaccines (GAP) aims to increase the production capacity of vaccines so that in the event of a pandemic there is an adequate supply to meet global needs. However, it has been estimated that even in the best case scenario there would be a considerable delay of at least five to six months for the first supplies of vaccine to become available after the isolation of the strain and availability of the candidate vaccine virus to vaccine manufacturers. By this time, the virus is likely to have already infected millions of people worldwide, causing significant mortality, morbidity and economic loss. Passive immunization through broadly neutralizing antibodies which bind to multiple, structurally diverse strains of influenza could be a promising solution to address the immediate health threat of an influenza pandemic while vaccines are being developed. These products may also have a role in seasonal influenza as an alternative to other options such as antivirals for the treatment of severe acute respiratory illness due to influenza. This article provides an overview of the current clinical pipeline of anti-influenza antibodies and discusses potential uses and the challenges to product development.
Collapse
Affiliation(s)
- Erin Sparrow
- The World Health Organization, Geneva, Switzerland; School of Public Health and Community Medicine, University of New South Wales, NSW, Australia.
| | | | - Mohamud Sheikh
- School of Public Health and Community Medicine, University of New South Wales, NSW, Australia
| | - Siranda Torvaldsen
- School of Public Health and Community Medicine, University of New South Wales, NSW, Australia; Sydney Medical School Northern, University of Sydney, NSW, Australia
| | - Anthony T Newall
- School of Public Health and Community Medicine, University of New South Wales, NSW, Australia
| |
Collapse
|