1
|
Li Q, Li H, Li Z, Wang Y. Vaccine and therapeutic agents against the respiratory syncytial virus: resolved and unresolved issue. MedComm (Beijing) 2024; 5:e70016. [PMID: 39575302 PMCID: PMC11581781 DOI: 10.1002/mco2.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a predominant pathogen responsible for respiratory tract infections among infants, the elderly, and immunocompromised individuals. In recent years, significant progress has been made in innovative vaccines and therapeutic agents targeting RSV. Nevertheless, numerous challenges and bottlenecks persist in the prevention and treatment of RSV infections. This review will provide an overview of the resolved and unresolved issues surrounding the development of vaccines and therapeutic agents against RSV. As of September 2024, three RSV vaccines against acute lower respiratory infections (ALRI) have been approved globally. Additionally, there have been notable progress in the realm of passive immunoprophylactic antibodies, with the monoclonal antibody nirsevimab receiving regulatory approval for the prevention of RSV infections in infants. Furthermore, a variety of RSV therapeutic agents are currently under clinical investigation, with the potential to yield breakthrough advancements in the foreseeable future. This review delineates the advancements and challenges faced in vaccines and therapeutic agents targeting RSV. It aims to provide insights that will guide the development of effective preventive and control measures for RSV.
Collapse
Affiliation(s)
- Qianqian Li
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- State Key Laboratory of Respiratory Health and MultimorbidityInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College)Ministry of EducationInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Huan Li
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- State Key Laboratory of Respiratory Health and MultimorbidityInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College)Ministry of EducationInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Zhihua Li
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- State Key Laboratory of Respiratory Health and MultimorbidityInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College)Ministry of EducationInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Youchun Wang
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- State Key Laboratory of Respiratory Health and MultimorbidityInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College)Ministry of EducationInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| |
Collapse
|
2
|
Kumar N, Sharma S, Kumar R, Meena VK, Barua S. Evolution of drug resistance against antiviral agents that target cellular factors. Virology 2024; 600:110239. [PMID: 39276671 DOI: 10.1016/j.virol.2024.110239] [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: 06/11/2024] [Revised: 07/29/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Antiviral drugs have classically been developed by directly disrupting the functions of viral proteins. However, this strategy has been largely unsuccessful due to the rapid generation of viral escape mutants. It has been well established that as compared to the virus-centric approach, the strategy of developing antiviral drugs by targeting host-dependency factors (HDFs) minimizes drug resistance. However, recent reports have indicated that drug resistance against some of the host-targeting antiviral agents can in fact occur under some circumstances. Long-term selection pressure of a host-targeting antiviral agent may induce the virus to use an alternate cellular factor or alters its affinity towards the target that confers resistance. Alternatively, virus may synchronize its life cycle with the patterns of drug therapy. In addition, virus may subvert host's immune system to perpetuate under the limiting conditions of the targeted cellular factor. This review describes novel potential mechanisms that may account for the acquiring resistance against agents that target HDFs.
Collapse
Affiliation(s)
- Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.
| | - Shalini Sharma
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Sciences, Sher-e-Kashmir University of Agricultural Sciences and Technology (SKAUST), Jammu, India.
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | | | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| |
Collapse
|
3
|
ElSherif M, Halperin SA. Benefits of Combining Molecular Biology and Controlled Human Infection Model Methodologies in Advancing Vaccine Development. J Mol Biol 2023; 435:168322. [PMID: 37866477 DOI: 10.1016/j.jmb.2023.168322] [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: 01/31/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Infectious diseases continue to account for a significant portion of global deaths despite the use of vaccines for several centuries. Immunization programs around the world are a testament to the great success of multiple vaccines, yet there are still diseases without vaccines and others that require safer more effective ones. Addressing uncontrolled and emerging disease threats is restrained by the limitations and bottlenecks encountered with traditional vaccine development paradigms. Recent advances in modern molecular biology technologies have enhanced the interrogation of host pathogen interaction and deciphered complex pathways, thereby uncovering the myriad interplay of biological events that generate immune protection against foreign agents. Consequent to insights into the immune system, modern biology has been instrumental in the development and production of next generation 21st century vaccines. As these biological tools, commonly and collectively referred to as 'omics, became readily available, there has been a renewed consideration of Controlled Human Infection Models (CHIMs). Successful and reproducible CHIMs can complement modern molecular biology for the study of infectious diseases and development of effective vaccines in a regulated process that mitigates risk, cost, and time, with capacity to discern immune correlates of protection.
Collapse
Affiliation(s)
- May ElSherif
- Canadian Center for Vaccinology, IWK Health, Nova Scotia Health, and Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Scott A Halperin
- Canadian Center for Vaccinology, IWK Health, Nova Scotia Health, and Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
4
|
Katzer M, Salloch S, Schindler C, Mertz M. Ethical Requirements for Human Challenge Studies: A Systematic Review of Reasons. Clin Pharmacol Ther 2023; 114:1209-1219. [PMID: 37716911 DOI: 10.1002/cpt.3054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Human challenge studies (HCS) are controlled clinical trials in which participants are deliberately infected with a pathogen. Such trials are being developed for an increasing number of diseases. Partly as a result of the coronavirus disease 2019 (COVID-19) pandemic, there has been a recent ethical debate about the reasons for and against HCS in general, or rather, about the requirements that individual HCS must fulfill to be ethically acceptable. A systematic review was conducted to categorize and summarize such requirements and the reasons given for them. Ethics literature was searched in PubMed, Google Scholar, BELIT, and PhilPapers; eligibility criteria were articles published in a scientific/scholarly journal (original research, reviews, editorials, opinion pieces, and conference/meeting reports). Of 1,322 records identified, 161 publications were included, with 183 requirements (with associated reasons) in 10 thematic categories extracted via qualitative content analysis. In synthesizing and interpreting the requirements and their reasons, three issues emerge as particularly sensitive in the case of HCS: the meaning of the right to withdraw from research procedures, communication of researchers with the public and various stakeholders, and the conditions of informed consent. However, four other issues, not specific to HCS, stand out as the most controversial: the acceptable level of risk to participants, payment of participants, protection of vulnerable groups, and standards for international collaborations. Controversies in these areas indicate that further debate is warranted, possibly leading to more specific instructions in ethics guidance documents.
Collapse
Affiliation(s)
- Matthias Katzer
- Institute for Ethics, History, and Philosophy of Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - Sabine Salloch
- Institute for Ethics, History, and Philosophy of Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - Christoph Schindler
- Center for Clinical Trials (ZKS), Early Clinical Trial Unit (ECTU) & Center for Pharmacology and Toxicology, Hannover Medical School (MHH), Hannover, Germany
| | - Marcel Mertz
- Institute for Ethics, History, and Philosophy of Medicine, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
5
|
Li Y, Wang X, Xu R, Wang T, Zhang D, Qian W. Establishment of RT-RPA-Cas12a assay for rapid and sensitive detection of human rhinovirus B. BMC Microbiol 2023; 23:333. [PMID: 37951882 PMCID: PMC10640725 DOI: 10.1186/s12866-023-03096-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 10/29/2023] [Indexed: 11/14/2023] Open
Abstract
Human rhinovirus B (HRV-B) is a major human viral pathogen that can be responsible for various kinds of infections. Due to the health risks associated with HRV-B, it is therefore crucial to explore a rapid, specific, and sensitive method for surveillance. Herein, we exploited a novel detection method for HRV-B by combining reverse-transcription recombinase polymerase amplification (RT-RPA) of nucleic acids isothermal amplification and the trans-cleavage activity of Cas12a. Our RT-RPA-Cas12a-based fluorescent assay can be completed within 35-45 min and obtain a lower detection threshold to 0.5 copies/µL of target RNA. Meanwhile, crRNA sequences without a specific protospacer adjacent motif can effectively activate the trans-cleavage activity of Cas12a. Moreover, our RT-RPA-Cas12a-based fluorescent method was examined using 30 clinical samples, and exhibited high accuracy with positive and negative predictive agreement of 90% and 100%, respectively. Taken together, a novel promising, rapid and effective RT-RPA-Cas12a-based detection method was explored and shows promising potential for on-site HRV-B infection in resource-limited settings.
Collapse
Affiliation(s)
- Yongdong Li
- Ningbo Key Laboratory of Virus Research, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, 315010, P. R. China
| | - Xuefei Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Rong Xu
- Ningbo Key Laboratory of Virus Research, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, 315010, P. R. China
| | - Ting Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Dandan Zhang
- Ningbo Key Laboratory of Virus Research, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, 315010, P. R. China.
| | - Weidong Qian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China.
| |
Collapse
|
6
|
Jordan E, Kabir G, Schultz S, Silbernagl G, Schmidt D, Jenkins VA, Weidenthaler H, Stroukova D, Martin BK, De Moerlooze L. Reduced Respiratory Syncytial Virus Load, Symptoms, and Infections: A Human Challenge Trial of MVA-BN-RSV Vaccine. J Infect Dis 2023; 228:999-1011. [PMID: 37079393 PMCID: PMC10582911 DOI: 10.1093/infdis/jiad108] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/20/2023] [Accepted: 04/19/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) causes significant disease burden in older adults. MVA-BN-RSV is a novel poxvirus-vectored vaccine encoding internal and external RSV proteins. METHODS In a phase 2a randomized double-blind, placebo-controlled trial, healthy participants aged 18 to 50 years received MVA-BN-RSV or placebo, then were challenged 4 weeks later with RSV-A Memphis 37b. Viral load was assessed from nasal washes. RSV symptoms were collected. Antibody titers and cellular markers were assessed before and after vaccination and challenge. RESULTS After receiving MVA-BN-RSV or placebo, 31 and 32 participants, respectively, were challenged. Viral load areas under the curve from nasal washes were lower (P = .017) for MVA-BN-RSV (median = 0.00) than placebo (median = 49.05). Total symptom scores also were lower (median = 2.50 and 27.00, respectively; P = .004). Vaccine efficacy against symptomatic, laboratory-confirmed or culture-confirmed infection was 79.3% to 88.5% (P = .022 and .013). Serum immunoglobulin A and G titers increased approximately 4-fold after MVA-BN-RSV vaccination. Interferon-γ-producing cells increased 4- to 6-fold after MVA-BN-RSV in response to stimulation with the encoded RSV internal antigens. Injection site pain occurred more frequently with MVA-BN-RSV. No serious adverse events were attributed to vaccination. CONCLUSIONS MVA-BN-RSV vaccination resulted in lower viral load and symptom scores, fewer confirmed infections, and induced humoral and cellular responses. CLINICAL TRIALS REGISTRATION NCT04752644.
Collapse
|
7
|
Ortiz JR, Bernstein DI, Hoft DF, Woods CW, McClain MT, Frey SE, Brady RC, Bryant C, Wegel A, Frenck RW, Walter EB, Abate G, Williams SR, Atmar RL, Keitel WA, Rouphael N, Memoli MJ, Makhene MK, Roberts PC, Neuzil KM. A Multicenter, Controlled Human Infection Study of Influenza A(H1N1)pdm09 in Healthy Adults. J Infect Dis 2023; 228:287-298. [PMID: 36702771 PMCID: PMC10420403 DOI: 10.1093/infdis/jiad021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND We evaluated the associations between baseline influenza virus-specific hemagglutination inhibition (HAI) and microneutralization (MN) titers and subsequent symptomatic influenza virus infection in a controlled human infection study. METHODS We inoculated unvaccinated healthy adults aged 18-49 years with an influenza A/California/04/2009/H1N1pdm-like virus (NCT04044352). We collected serial safety labs, serum for HAI and MN, and nasopharyngeal swabs for reverse-transcription polymerase chain reaction (RT-PCR) testing. Analyses used the putative seroprotective titer of ≥40 for HAI and MN. The primary clinical outcome was mild-to-moderate influenza disease (MMID), defined as ≥1 postchallenge positive qualitative RT-PCR test with a qualifying symptom/clinical finding. RESULTS Of 76 participants given influenza virus challenge, 54 (71.1%) experienced MMID. Clinical illness was generally very mild. MMID attack rates among participants with baseline titers ≥40 by HAI and MN were 64.9% and 67.9%, respectively, while MMID attack rates among participants with baseline titers <40 by HAI and MN were 76.9% and 78.3%, respectively. The estimated odds of developing MMID decreased by 19% (odds ratio, 0.81 [95% confidence interval, .62-1.06]; P = .126) for every 2-fold increase in baseline HAI. There were no significant adverse events. CONCLUSIONS We achieved a 71.1% attack rate of MMID. High baseline HAI and MN were associated with protection from illness. Clinical Trials Registration. NCT04044352.
Collapse
Affiliation(s)
- Justin R Ortiz
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - David I Bernstein
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Daniel F Hoft
- Internal Medicine and
- Molecular Microbiology and Immunology, Division of Infectious Diseases, Allergy and Immunology and Center for Vaccine Development, Saint Louis University School of Medicine, Missouri
| | - Christopher W Woods
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | - Micah T McClain
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | | | - Rebecca C Brady
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Christopher Bryant
- Vaccine and Infectious Disease Therapeutic Research Unit, The Emmes Company, Rockville, Maryland
| | - Ashley Wegel
- Vaccine and Infectious Disease Therapeutic Research Unit, The Emmes Company, Rockville, Maryland
| | - Robert W Frenck
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | | | - Sarah R Williams
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore
| | - Robert L Atmar
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Wendy A Keitel
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | | | - Mamodikoe K Makhene
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
8
|
von Delft A, Hall MD, Kwong AD, Purcell LA, Saikatendu KS, Schmitz U, Tallarico JA, Lee AA. Accelerating antiviral drug discovery: lessons from COVID-19. Nat Rev Drug Discov 2023; 22:585-603. [PMID: 37173515 PMCID: PMC10176316 DOI: 10.1038/s41573-023-00692-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, a wave of rapid and collaborative drug discovery efforts took place in academia and industry, culminating in several therapeutics being discovered, approved and deployed in a 2-year time frame. This article summarizes the collective experience of several pharmaceutical companies and academic collaborations that were active in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antiviral discovery. We outline our opinions and experiences on key stages in the small-molecule drug discovery process: target selection, medicinal chemistry, antiviral assays, animal efficacy and attempts to pre-empt resistance. We propose strategies that could accelerate future efforts and argue that a key bottleneck is the lack of quality chemical probes around understudied viral targets, which would serve as a starting point for drug discovery. Considering the small size of the viral proteome, comprehensively building an arsenal of probes for proteins in viruses of pandemic concern is a worthwhile and tractable challenge for the community.
Collapse
Affiliation(s)
- Annette von Delft
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, National Institute for Health Research, University of Oxford, Oxford, UK.
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | | | | | | | | | | | - Alpha A Lee
- PostEra, Inc., Cambridge, MA, USA.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Işık YE, Aydın Z. Comparative analysis of machine learning approaches for predicting respiratory virus infection and symptom severity. PeerJ 2023; 11:e15552. [PMID: 37404475 PMCID: PMC10317018 DOI: 10.7717/peerj.15552] [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: 11/11/2022] [Accepted: 05/23/2023] [Indexed: 07/06/2023] Open
Abstract
Respiratory diseases are among the major health problems causing a burden on hospitals. Diagnosis of infection and rapid prediction of severity without time-consuming clinical tests could be beneficial in preventing the spread and progression of the disease, especially in countries where health systems remain incapable. Personalized medicine studies involving statistics and computer technologies could help to address this need. In addition to individual studies, competitions are also held such as Dialogue for Reverse Engineering Assessment and Methods (DREAM) challenge which is a community-driven organization with a mission to research biology, bioinformatics, and biomedicine. One of these competitions was the Respiratory Viral DREAM Challenge, which aimed to develop early predictive biomarkers for respiratory virus infections. These efforts are promising, however, the prediction performance of the computational methods developed for detecting respiratory diseases still has room for improvement. In this study, we focused on improving the performance of predicting the infection and symptom severity of individuals infected with various respiratory viruses using gene expression data collected before and after exposure. The publicly available gene expression dataset in the Gene Expression Omnibus, named GSE73072, containing samples exposed to four respiratory viruses (H1N1, H3N2, human rhinovirus (HRV), and respiratory syncytial virus (RSV)) was used as input data. Various preprocessing methods and machine learning algorithms were implemented and compared to achieve the best prediction performance. The experimental results showed that the proposed approaches obtained a prediction performance of 0.9746 area under the precision-recall curve (AUPRC) for infection (i.e., shedding) prediction (SC-1), 0.9182 AUPRC for symptom class prediction (SC-2), and 0.6733 Pearson correlation for symptom score prediction (SC-3) by outperforming the best leaderboard scores of Respiratory Viral DREAM Challenge (a 4.48% improvement for SC-1, a 13.68% improvement for SC-2, and a 13.98% improvement for SC-3). Additionally, over-representation analysis (ORA), which is a statistical method for objectively determining whether certain genes are more prevalent in pre-defined sets such as pathways, was applied using the most significant genes selected by feature selection methods. The results show that pathways associated with the 'adaptive immune system' and 'immune disease' are strongly linked to pre-infection and symptom development. These findings contribute to our knowledge about predicting respiratory infections and are expected to facilitate the development of future studies that concentrate on predicting not only infections but also the associated symptoms.
Collapse
Affiliation(s)
- Yunus Emre Işık
- Department of Management Information Systems, Sivas Cumhuriyet University, Sivas, Turkey
| | - Zafer Aydın
- Department of Computer Engineering, Abdullah Gül University, Kayseri, Turkey
| |
Collapse
|
10
|
The WE SENSE study protocol: A controlled, longitudinal clinical trial on the use of wearable sensors for early detection and tracking of viral respiratory tract infections. Contemp Clin Trials 2023; 128:107103. [PMID: 37147083 PMCID: PMC10049920 DOI: 10.1016/j.cct.2023.107103] [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: 08/08/2022] [Revised: 12/06/2022] [Accepted: 01/20/2023] [Indexed: 03/31/2023]
Abstract
Background Viral respiratory tract infections (VRTI) are extremely common. Considering the profound social and economic impact of COVID-19, it is imperative to identify novel mechanisms for early detection and prevention of VRTIs, to prevent future pandemics. Wearable biosensor technology may facilitate this. Early asymptomatic detection of VRTIs could reduce stress on the healthcare system by reducing transmission and decreasing the overall number of cases. The aim of the current study is to define a sensitive set of physiological and immunological signature patterns of VRTI through machine learning (ML) to analyze physiological data collected continuously using wearable vital signs sensors. Methods A controlled, prospective longitudinal study with an induced low grade viral challenge, coupled with 12 days of continuous wearable biosensors monitoring surrounding viral induction. We aim to recruit and simulate a low grade VRTI in 60 healthy adults aged 18–59 years via administration of live attenuated influenza vaccine (LAIV). Continuous monitoring with wearable biosensors will include 7 days pre (baseline) and 5 days post LAIV administration, during which vital signs and activity-monitoring biosensors (embedded in a shirt, wristwatch and ring) will continuously monitor physiological and activity parameters. Novel infection detection techniques will be developed based on inflammatory biomarker mapping, PCR testing, and app-based VRTI symptom tracking. Subtle patterns of change will be assessed via ML algorithms developed to analyze large datasets and generate a predictive algorithm. Conclusion This study presents an infrastructure to test wearables for the detection of asymptomatic VRTI using multimodal biosensors, based on immune host response signature. CliniclTrials.govregistration:NCT05290792
Collapse
|
11
|
McKay S, Teitsma-Jansen A, Floris E, Dekker T, Smids B, Khurshid R, Calame W, Kardinaal A, Lutter R, Albers R. Effects of Dietary Supplementation with Carrot-Derived Rhamnogalacturonan-I (cRG-I) on Accelerated Protective Immune Responses and Quality of Life in Healthy Volunteers Challenged with Rhinovirus in a Randomized Trial. Nutrients 2022; 14:4258. [PMID: 36296939 PMCID: PMC9607575 DOI: 10.3390/nu14204258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
An adequate and balanced supply of nutrients is essential for maintaining health, and an optimal immune response is fast, contained and properly controlled, curbing infections quickly while minimizing damage. Several micronutrients contribute to normal immune function and certain dietary fibers, for example pectic polysaccharides, can play an important role in educating and regulating immune cell responses. The aim of this paper is to elaborate on our initial findings that dietary supplementation with carrot-derived rhamnogalacturonan-I (cRG-I) accelerates and augments local innate immune and anti-viral interferon response to a rhinovirus-16 (RV16) infection and reduces the severity and duration of symptoms in humans. Dietary intake of cRG-I also enhanced immune responses to this respiratory viral infection as measured by ex vivo stimulation of whole blood with the Toll-like receptor 3 (TLR3) ligand polyinosinic:polycytidylic acid and NK cell function. Consumption of cRG-I also reduced the negative effects of this common cold infection on quality of life as assessed by individual symptom scores. RG-I from carrot is a safe, sustainable, and economically viable solution that could easily be integrated into food products and dietary supplements aiming to support immune fitness and wellbeing.
Collapse
Affiliation(s)
- Sue McKay
- NutriLeads B.V., Bronland 12-N, 6708 WH Wageningen, The Netherlands
| | - Annemarie Teitsma-Jansen
- Amsterdam UMC, Department of Experimental Immunology, University of Amsterdam and Amsterdam Infection & Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | | | - Tamara Dekker
- Amsterdam UMC, Department of Experimental Immunology, University of Amsterdam and Amsterdam Infection & Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Barbara Smids
- Amsterdam UMC, Department of Experimental Immunology, University of Amsterdam and Amsterdam Infection & Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ridha Khurshid
- Amsterdam UMC, Department of Experimental Immunology, University of Amsterdam and Amsterdam Infection & Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Wim Calame
- StatistiCal B.V., Strandwal 148, 2241 MN Wassenaar, The Netherlands
| | | | - René Lutter
- Amsterdam UMC, Department of Experimental Immunology, University of Amsterdam and Amsterdam Infection & Immunity Institute, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ruud Albers
- NutriLeads B.V., Bronland 12-N, 6708 WH Wageningen, The Netherlands
| |
Collapse
|
12
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
13
|
Lim DK, Kim JW, Kim JK. Effects of climatic factors on the prevalence of influenza virus infection in Cheonan, Korea. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:59052-59059. [PMID: 35381925 DOI: 10.1007/s11356-022-20070-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Big data can be used to correlate diseases and climatic factors. The prevalence of influenza (flu) virus, accounting for a large proportion of respiratory infections, suggests that the effect of climate variables according to seasonal dynamics of influenza virus infections should be investigated. Here, trends in flu virus detection were analyzed using data from 9,010 tests performed between January 2012 and December 2018 at Dankook University Hospital, Cheonan, Korea. We compared the detection of the flu virus in Cheonan area and its association with climate change. The flu virus detection rate was 9.9% (894/9,010), and the detection rate was higher for flu virus A (FLUAV; 6.9%) than for flu virus B (FLUBV; 3.0%). Both FLUAV and FLUBV infections are considered an epidemic each year. We identified 43.1% (n = 385) and 35.0% (n = 313) infections in children aged < 10 years and adults aged > 60 years, respectively. The combination of these age groups encompassed 78.1% (n = 698/894) of the total data. Flu virus infections correlated with air temperature, relative humidity, vapor pressure, atmospheric pressure, particulate matter, and wind chill temperature (P < 0.001). However, the daily temperature range did not significantly correlate with the flu detection results. This is the first study to identify the relationship between long-term flu virus infection with temperature in the temperate region of Cheonan.
Collapse
Affiliation(s)
- Dong Kyu Lim
- Department of Medical Laser, Dankook University Graduate School of Medicine, Chungnam, South Korea
| | - Jong Wan Kim
- Department of Laboratory Medicine, Dankook University College of Medicine, Chungnam, South Korea
| | - Jae Kyung Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, South Korea.
| |
Collapse
|
14
|
Medina MJ, Nazareth J, Dillon HM, Wighton CJ, Bandi S, Pan D, Nicholson KG, Clark TW, Andrew PW, Pareek M. Respiratory virus transmission using a novel viral challenge model: an observational cohort study. J Infect 2022; 85:405-411. [DOI: 10.1016/j.jinf.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
|
15
|
Lim DK, Jeon JS, Jang TS, Kim JK. Association between climatic factors and respiratory syncytial virus detection rates in Cheonan, Korea. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:13315-13322. [PMID: 34585349 DOI: 10.1007/s11356-021-16546-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/10/2021] [Indexed: 06/13/2023]
Abstract
The use of big data may facilitate the recognition and interpretation of causal relationships between disease occurrence and climatic variables. This study examined the effects of various climatic variables on the seasonal epidemiology of respiratory syncytial virus (RSV) infections in the temperate climate of Korea. Trends in RSV detection were analyzed using 9010 samples tested between January 1, 2012, and December 31, 2018, at Dankook University Hospital in Cheonan, Korea. Seasonal patterns in RSV detection frequency were compared with local climatic variables during the same period. RSV detection rate of 12.8% (n = 1150/9010) was observed, which was higher for RSV-A (7.1%) than RSV-B (5.8%) and RSV-A and RSV-B alternated each year. Children < 1 year exhibited high infection rates with RSV-A (68.5%) and RSV-B (58.7%). RSV-A and RSV-B infection rates in children under 9 years old were 96.2% and 92.1%, respectively. RSV had a significant relationship with several climatic factors. Air temperature, wind chill temperature, and particulate matter concentration were lower on days with a higher frequency of RSV detection. In contrast, atmospheric pressure was higher on days with lower RSV detection. Although the detection rates for RSV-A and RSV-B increased on days with lower air temperatures, those for RSV-B also increased on days with lower wind chill temperatures. Our findings suggest that climatic variables affect the RSV detection rate among children under 10 years of age. The present data may help predict the time when prevention strategies may be the most effective.
Collapse
Affiliation(s)
- Dong Kyu Lim
- Department of Medical Laser, Dankook University Graduate School of Medicine, Cheonan-si, South Korea
| | - Jae-Sik Jeon
- Department of Biomedical Laboratory Science, College of Health Sciences, Dankook University, Cheonan-si, South Korea
| | - Tae Su Jang
- Department of Medicine, College of Medicine, Dankook University, Cheonan-si, South Korea
| | - Jae Kyung Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Dankook University, Cheonan-si, South Korea.
| |
Collapse
|
16
|
Turner RB, Lehtoranta L, Hibberd A, Männikkö S, Zabel B, Yeung N, Huttunen T, Burns FR, Lehtinen MJ. Effect of Bifidobacterium animalis spp. lactis Bl-04 on Rhinovirus-Induced Colds: A Randomized, Placebo-Controlled, Single-Center, Phase II Trial in Healthy Volunteers. EClinicalMedicine 2022; 43:101224. [PMID: 34927036 PMCID: PMC8649651 DOI: 10.1016/j.eclinm.2021.101224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND This study was designed to assess the efficacy of Bifidobacterium animalis ssp. lactis (Bl-04) for prevention of rhinovirus colds and to explore the interactions between the probiotic, the viral infection, the host response and the host microbiome. METHODS The effect of ingestion of the probiotic Bl-04 was evaluated in a randomized, double-blinded rhinovirus (RV) challenge study. Healthy volunteers recruited from a university community in USA were randomized 1:1 using a computer generated code to ingest either Bl-04 (n=165) or placebo (n=169) for 28 days and were then challenged with RV-A39, and followed for 14 days. All study interactions and sample collection occurred in dedicated clinical research space. The primary analysis was the effect of the probiotic on the incidence of RV-associated illness. (Trial registration: NCT02679807, study complete). FINDINGS The first cohort of volunteers was randomized on March 14, 2016 and the last (5th) cohort was randomized on March 12, 2018. Sixty-three (56%, 95% CI [47%; 66%]) of the 112 subjects in the active group and 60 (50%,95% CI [41%; 59%]) of the 120 subjects in the placebo group had a protocol-defined rhinovirus-associated illness (χ2=0·91, p=0·34). The point estimate of the difference in illness (active-placebo) is 6.3% (95% CI -6.7;19.1). There were no adverse events that were judged as definitely or probably related to the study product. INTERPRETATION In this study there was no effect of orally administered Bl-04 on the occurrence of RV-associated illness. FUNDING Danisco Sweeteners Oy (now IFF Health & Biosciences).
Collapse
Affiliation(s)
- Ronald B. Turner
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
- Corresponding To: 512 Rosemont Drive, Charlottesville, VA 22903
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Woodall MNJ, Masonou T, Case K, Smith CM. Human models for COVID-19 research. J Physiol 2021; 599:4255-4267. [PMID: 34287894 PMCID: PMC8447334 DOI: 10.1113/jp281499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Currently, therapeutics for COVID-19 are limited. To overcome this, it is important that we use physiologically relevant models to reproduce the pathology of infection and evaluate the efficacy of antiviral drugs. Models of airway infection, including the use of a human infection challenge model or well-defined, disease relevant in vitro systems can help determine the key components that perpetuate the severity of the disease. Here, we briefly review the human models that are currently being used in COVID-19 research and drug development.
Collapse
Affiliation(s)
| | - Tereza Masonou
- GOS Institute of Child HealthUniversity College LondonLondonUK
| | | | - Claire M. Smith
- GOS Institute of Child HealthUniversity College LondonLondonUK
| |
Collapse
|
18
|
Rajan A, Weaver AM, Aloisio GM, Jelinski J, Johnson HL, Venable SF, McBride T, Aideyan L, Piedra FA, Ye X, Melicoff-Portillo E, Yerramilli MRK, Zeng XL, Mancini MA, Stossi F, Maresso AW, Kotkar SA, Estes MK, Blutt S, Avadhanula V, Piedra PA. The human nose organoid respiratory virus model: an ex-vivo human challenge model to study RSV and SARS-CoV-2 pathogenesis and evaluate therapeutics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34341793 DOI: 10.1101/2021.07.28.453844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
There is an unmet need for pre-clinical models to understand the pathogenesis of human respiratory viruses; and predict responsiveness to immunotherapies. Airway organoids can serve as an ex-vivo human airway model to study respiratory viral pathogenesis; however, they rely on invasive techniques to obtain patient samples. Here, we report a non-invasive technique to generate human nose organoids (HNOs) as an alternate to biopsy derived organoids. We made air liquid interface (ALI) cultures from HNOs and assessed infection with two major human respiratory viruses, respiratory syncytial virus (RSV) and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Infected HNO-ALI cultures recapitulate aspects of RSV and SARS-CoV-2 infection, including viral shedding, ciliary damage, innate immune responses, and mucus hyper-secretion. Next, we evaluated the feasibility of the HNO-ALI respiratory virus model system to test the efficacy of palivizumab to prevent RSV infection. Palivizumab was administered in the basolateral compartment (circulation) while viral infection occurred in the apical ciliated cells (airways), simulating the events in infants. In our model, palivizumab effectively prevented RSV infection in a concentration dependent manner. Thus, the HNO-ALI model can serve as an alternate to lung organoids to study respiratory viruses and testing therapeutics.
Collapse
|
19
|
Animal Models Utilized for the Development of Influenza Virus Vaccines. Vaccines (Basel) 2021; 9:vaccines9070787. [PMID: 34358203 PMCID: PMC8310120 DOI: 10.3390/vaccines9070787] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/25/2022] Open
Abstract
Animal models have been an important tool for the development of influenza virus vaccines since the 1940s. Over the past 80 years, influenza virus vaccines have evolved into more complex formulations, including trivalent and quadrivalent inactivated vaccines, live-attenuated vaccines, and subunit vaccines. However, annual effectiveness data shows that current vaccines have varying levels of protection that range between 40–60% and must be reformulated every few years to combat antigenic drift. To address these issues, novel influenza virus vaccines are currently in development. These vaccines rely heavily on animal models to determine efficacy and immunogenicity. In this review, we describe seasonal and novel influenza virus vaccines and highlight important animal models used to develop them.
Collapse
|
20
|
Satia I, Adatia A, Cusack RP, Greene JM, O'Byrne PM, Killian KJ, Johnston N. Influence of age, sex and respiratory viruses on the rates of emergency department visits and hospitalisations with respiratory tract infections, asthma and COPD. ERJ Open Res 2021; 7:00053-2021. [PMID: 34046485 PMCID: PMC8141702 DOI: 10.1183/23120541.00053-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/18/2021] [Indexed: 01/08/2023] Open
Abstract
Background The importance of age, sex and respiratory virus prevalence in emergency department (ED) visits and hospitalisations for respiratory tract infections (RTIs), asthma and COPD in a whole population over time is not well established. Methods This study retrospectively analysed data for daily ED visits and hospitalisations from 2003 to 2013 in Ontario, Canada and the daily number of virus positive tests. Daily numbers of ED visits and hospitalisations with RTIs, asthma and COPD listed as a primary diagnosis were collected from the Canadian Institute for Health Information. Virus data were obtained from the Respiratory Virus Detection Surveillance System. Multiple linear regression was used to assess the association of individual viruses with the daily rates. Results There were 4 365 578 ED visits and 321 719 (7.4%) admissions for RTIs, 817 141 ED visits and 260 665 (31.9%) admissions for COPD and 649 666 ED visits and 68 626 (10.6%) admissions for asthma. Respiratory syncytial virus and influenza A were associated with male ED visits, whereas human rhinovirus was associated with female ED visits for RTIs in preschool children. 19.2% of males, but only 7.2% of females were admitted. The correlation between the prevalence of each virus and ED visits and hospitalisations for asthma was weak, irrespective of age group and sex. Influenza A was most strongly associated with COPD ED visits and hospitalisations in males and females. Conclusions There are significant age and sex differences in the contribution of respiratory viruses to the number of ED visits and hospitalisations for RTIs, asthma and COPD. There are important age- and sex-related differences in the contribution of respiratory viruses to the number of ED visits and hospitalisations for respiratory tract infections, asthma and COPDhttps://bit.ly/39hrhIW
Collapse
Affiliation(s)
- Imran Satia
- Dept of Medicine, McMaster University, Hamilton, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Canada.,Division of Infection, Immunity and Respiratory Medicine, and Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,These authors contributed equally
| | - Adil Adatia
- Dept of Medicine, McMaster University, Hamilton, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Canada.,These authors contributed equally
| | - Ruth P Cusack
- Dept of Medicine, McMaster University, Hamilton, Canada
| | | | - Paul M O'Byrne
- Dept of Medicine, McMaster University, Hamilton, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Canada
| | | | - Neil Johnston
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Canada
| |
Collapse
|
21
|
Lynch HF, Darton TC, Levy J, McCormick F, Ogbogu U, Payne RO, Roth AE, Shah AJ, Smiley T, Largent EA. Promoting Ethical Payment in Human Infection Challenge Studies. THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2021; 21:11-31. [PMID: 33541252 DOI: 10.1080/15265161.2020.1854368] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
To prepare for potential human infection challenge studies (HICS) involving SARS-CoV-2, we convened a multidisciplinary working group to address ethical questions regarding whether and how much SARS-CoV-2 HICS participants should be paid. Because the goals of paying HICS participants, as well as the relevant ethical concerns, are the same as those arising for other types of clinical research, the same basic framework for ethical payment can apply. This framework divides payment into reimbursement, compensation, and incentives, focusing on fairness and promoting adequate recruitment and retention as counterweights to concerns about undue inducement. Within the basic framework, several factors are especially salient for HICS, and for SARS-CoV-2 HICS in particular, including the nature of participant confinement, anticipated discomfort, risks and uncertainty, participant motivations, and trust. These factors are reflected in a payment worksheet created to help sponsors, researchers, and ethics reviewers systematically develop and assess ethically justifiable payment amounts.
Collapse
|
22
|
Rajan A, Weaver AM, Aloisio GM, Jelinski J, Johnson HL, Venable SF, McBride T, Aideyan L, Piedra FA, Ye X, Melicoff-Portillo E, Yerramilli MRK, Zeng XL, Mancini MA, Stossi F, Maresso AW, Kotkar SA, Estes MK, Blutt S, Avadhanula V, Piedra PA. The Human Nose Organoid Respiratory Virus Model: an Ex Vivo Human Challenge Model To Study Respiratory Syncytial Virus (RSV) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Pathogenesis and Evaluate Therapeutics. mBio 2021; 13:e0351121. [PMID: 35164569 PMCID: PMC8844923 DOI: 10.1128/mbio.03511-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
There is an unmet need for preclinical models to understand the pathogenesis of human respiratory viruses and predict responsiveness to immunotherapies. Airway organoids can serve as an ex vivo human airway model to study respiratory viral pathogenesis; however, they rely on invasive techniques to obtain patient samples. Here, we report a noninvasive technique to generate human nose organoids (HNOs) as an alternative to biopsy-derived organoids. We made air-liquid interface (ALI) cultures from HNOs and assessed infection with two major human respiratory viruses, respiratory syncytial virus (RSV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Infected HNO-ALI cultures recapitulate aspects of RSV and SARS-CoV-2 infection, including viral shedding, ciliary damage, innate immune responses, and mucus hypersecretion. Next, we evaluated the feasibility of the HNO-ALI respiratory virus model system to test the efficacy of palivizumab to prevent RSV infection. Palivizumab was administered in the basolateral compartment (circulation), while viral infection occurred in the apical ciliated cells (airways), simulating the events in infants. In our model, palivizumab effectively prevented RSV infection in a concentration-dependent manner. Thus, the HNO-ALI model can serve as an alternative to lung organoids to study respiratory viruses and test therapeutics. IMPORTANCE Preclinical models that recapitulate aspects of human airway disease are essential for the advancement of novel therapeutics and vaccines. Here, we report a versatile airway organoid model, the human nose organoid (HNO), that recapitulates the complex interactions between the host and virus. HNOs are obtained using noninvasive procedures and show divergent responses to SARS-CoV-2 and RSV infection. SARS-CoV-2 induces severe damage to cilia and the epithelium, no interferon-λ response, and minimal mucus secretion. In striking contrast, RSV induces hypersecretion of mucus and a profound interferon-λ response with ciliary damage. We also demonstrated the usefulness of our ex vivo HNO model of RSV infection to test the efficacy of palivizumab, an FDA-approved monoclonal antibody to prevent severe RSV disease in high-risk infants. Our study reports a breakthrough in both the development of a novel nose organoid model and in our understanding of the host cellular response to RSV and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Ashley Morgan Weaver
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Gina Marie Aloisio
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph Jelinski
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Hannah L. Johnson
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Susan F. Venable
- Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Trevor McBride
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Letisha Aideyan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Felipe-Andrés Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Xunyan Ye
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | - Xi-Lei Zeng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael A. Mancini
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Fabio Stossi
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Shalaka A. Kotkar
- Environmental Safety Department, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Section of Infectious Diseases and Gastroenterology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
23
|
Kirchhelle C, Vanderslott S. Editorial: The Need for Harmonised International Guidelines ahead of COVID-19 Human Infection Studies. Public Health Rev 2021; 42:1603962. [PMID: 33796341 PMCID: PMC7904247 DOI: 10.3389/phrs.2021.1603962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/12/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Claas Kirchhelle
- University College Dublin, Dublin, United Kingdom
- Oxford Martin School, University of Oxford, Oxford, United Kingdom
| | - Samantha Vanderslott
- Oxford Vaccine Group and Oxford Martin School, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
A fully automated microfluidic PCR-array system for rapid detection of multiple respiratory tract infection pathogens. Anal Bioanal Chem 2021; 413:1787-1798. [PMID: 33492406 PMCID: PMC7829496 DOI: 10.1007/s00216-021-03171-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022]
Abstract
Rapid and accurate identification of respiratory tract infection pathogens is of utmost importance for clinical diagnosis and treatment, as well as prevention of pathogen transmission. To meet this demand, a microfluidic chip-based PCR-array system, Onestart, was developed. The Onestart system uses a microfluidic chip packaged with all the reagents required, and the waste liquid is also collected and stored on the chip. This ready-to-use system can complete the detection of 21 pathogens in a fully integrated manner, with sample lysis, nucleic acid extraction/purification, and real-time PCR sequentially implemented on the same chip. The entire analysis process is completed within 1.5 h, and the system automatically generates a test report. The lower limit-of-detection (LOD) of the Onestart assay was determined to be 1.0 × 103 copies·mL−1. The inter-batch variation of cycle threshold (Ct) values ranged from 0.08% to 0.69%, and the intra-batch variation ranged from 0.9% to 2.66%. Analytical results of the reference sample mix showed a 100% specificity of the Onestart assay. The analysis of batched clinical samples showed consistency of the Onestart assay with real-time PCR. With its ability to provide rapid, sensitive, and specific detection of respiratory tract infection pathogens, application of the Onestart system will facilitate timely clinical management of respiratory tract infections and effective prevention of pathogen transmission.
Collapse
|
25
|
Abstract
Viral infections and their emergence continue to pose a threat to human lives. Up to the present, there are limited numbers of vaccines that effectively work and few antivirals licensed for use in clinical practice. Added to this is the increase in antiviral resistance, meaning that drugs that do work are at risk of reduced efficacy. The recent global pandemic of coronavirus 2019 has provided evidence for the risk of a preventative vaccination and effective treatment of viruses' subsequent consequences. The aim of this article is to review traditional and herbal treatments for infections, specifically addressing gastrointestinal and respiratory viral infections.
Collapse
|
26
|
Lambkin‐Williams R, DeVincenzo JP. A COVID-19 human viral challenge model. Learning from experience. Influenza Other Respir Viruses 2020; 14:747-756. [PMID: 32790065 PMCID: PMC7578316 DOI: 10.1111/irv.12797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 01/15/2023] Open
Abstract
The controlled human infection model and specifically the human viral challenge model are not dissimilar to standard clinical trials while adding another layer of complexity and safety considerations. The models deliberately infect volunteers, with an infectious challenge agent to determine the effect of the infection and the potential benefits of the experimental interventions. The human viral challenge model studies can shorten the time to assess the efficacy of a new vaccine or treatment by combining this with the assessment of safety. The newly emerging SARS-CoV-2 virus is highly contagious, and an urgent race is on to develop a new vaccine against this virus in a timeframe never attempted before. The use of the human viral challenge model has been proposed to accelerate the development of the vaccine. In the early 2000s, the authors successfully developed a pathogenic human viral challenge model for another virus for which there was no effective treatment and established it to evaluate potential therapies and vaccines against respiratory syncytial virus. Experience gained in the development of that model can help with the development of a COVID-19 HVCM and the authors describe it here.
Collapse
|
27
|
Kim JM, Jeon JS, Kim JK. Climate and Human coronaviruses 229E and Human coronaviruses OC43 Infections: Respiratory Viral Infections Prevalence in Hospitalized Children in Cheonan, Korea. J Microbiol Biotechnol 2020; 30:1495-1499. [PMID: 32807752 PMCID: PMC9728399 DOI: 10.4014/jmb.2004.04052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/13/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022]
Abstract
The study of climate and respiratory viral infections using big data may enable the recognition and interpretation of relationships between disease occurrence and climatic variables. In this study, realtime reverse transcription quantitative PCR (qPCR) methods were used to identify Human respiratory coronaviruses (HCoV). infections in patients below 10 years of age with respiratory infections who visited Dankook University Hospital in Cheonan, South Korea, from January 1, 2012, to December 31, 2018. Out of the 9010 patients who underwent respiratory virus real-time reverse transcription qPCR test, 364 tested positive for HCoV infections. Among these 364 patients, 72.8% (n = 265) were below 10 years of age. Data regarding the frequency of infections was used to uncover the seasonal pattern of the two viral strains, which was then compared with local meteorological data for the same time period. HCoV-229E and HCoV-OC43 showed high infection rates in patients below 10 years of age. There was a negative relationship between HCoV-229E and HCoV-OC43 infections with air temperature and wind-chill temperatures. Both HCoV-229E and HCoV-OC43 rates of infection were positively related to atmospheric pressure, while HCoV-229E was also positively associated with particulate matter concentrations. Our results suggest that climatic variables affect the rate in which children below 10 years of age are infected with HCoV. These findings may help to predict when prevention strategies may be most effective.
Collapse
Affiliation(s)
- Jang Mook Kim
- Department of Health Administration, College of Health Sciences, Dankook University, Cheonan 31116, Republic of Korea
| | - Jae Sik Jeon
- Department of Biomedical Laboratory Science, College of Health Sciences, Dankook University, Cheonan 31116, Republic of Korea
| | - Jae Kyung Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Dankook University, Cheonan 31116, Republic of Korea,Corresponding author Phone: +82-41-550-1451 Fax: +82-41-559-7934 E-mail:
| |
Collapse
|
28
|
Investigation of Plasmid DNA Delivery and Cell Viability Dynamics for Optimal Cell Electrotransfection In Vitro. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10176070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Electroporation is an effective method for delivering plasmid DNA molecules into cells. The efficiency of gene electrotransfer depends on several factors. To achieve high transfection efficiency while maintaining cell viability is a tedious task in electroporation. Here, we present a combined study in which the dynamics of both evaluation types of transfection efficiency and the cell viability were evaluated in dependence of plasmid concentration as well as at the different number of high voltage (HV) electric pulses. The results of this study reveal a quantitative sigmoidal (R2 > 0.95) dependence of the transfection efficiency and cell viability on the distance between the cell membrane and the nearest plasmid. We propose this distance value as a new, more accurate output parameter that could be used in further optimization studies as a predictor and a measure of electrotransfection efficiency.
Collapse
|
29
|
Next Generation Influenza Vaccines: Looking into the Crystal Ball. Vaccines (Basel) 2020; 8:vaccines8030464. [PMID: 32825635 PMCID: PMC7563445 DOI: 10.3390/vaccines8030464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 11/29/2022] Open
Abstract
Influenza infections are responsible for significant number of deaths and overwhelming costs worldwide every year. Vaccination represents the only cost-efficient alternative to address this major problem in human health. However, current vaccines are fraught by many limitations, being far from optimal. Among them, the need to upgrade vaccines every year through a time-consuming process open to different caveats, and the critical fact that they exhibit poorer efficacy in individuals who are at high risk for severe infections. Where are we? How can knowledge and technologies contribute towards removing current roadblocks? What does the future offer in terms of next generation vaccines?
Collapse
|
30
|
|
31
|
Aggarwal M, Plemper RK. Structural Insight into Paramyxovirus and Pneumovirus Entry Inhibition. Viruses 2020; 12:E342. [PMID: 32245118 PMCID: PMC7150754 DOI: 10.3390/v12030342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 01/04/2023] Open
Abstract
Paramyxoviruses and pneumoviruses infect cells through fusion (F) protein-mediated merger of the viral envelope with target membranes. Members of these families include a range of major human and animal pathogens, such as respiratory syncytial virus (RSV), measles virus (MeV), human parainfluenza viruses (HPIVs), and highly pathogenic Nipah virus (NiV). High-resolution F protein structures in both the metastable pre- and the postfusion conformation have been solved for several members of the families and a number of F-targeting entry inhibitors have progressed to advanced development or clinical testing. However, small-molecule RSV entry inhibitors have overall disappointed in clinical trials and viral resistance developed rapidly in experimental settings and patients, raising the question of whether the available structural information may provide a path to counteract viral escape through proactive inhibitor engineering. This article will summarize current mechanistic insight into F-mediated membrane fusion and examine the contribution of structural information to the development of small-molecule F inhibitors. Implications are outlined for future drug target selection and rational drug engineering strategies.
Collapse
Affiliation(s)
| | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA;
| |
Collapse
|
32
|
Parčina M, Schneider UV, Visseaux B, Jozić R, Hannet I, Lisby JG. Multicenter evaluation of the QIAstat Respiratory Panel-A new rapid highly multiplexed PCR based assay for diagnosis of acute respiratory tract infections. PLoS One 2020; 15:e0230183. [PMID: 32163484 PMCID: PMC7067435 DOI: 10.1371/journal.pone.0230183] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 02/24/2020] [Indexed: 01/06/2023] Open
Abstract
Acute respiratory tract infections (ARTI), including the common cold, pharyngitis, sinusitis, otitis media, bronchiolitis and pneumonia are the most common diagnoses among patients seeking medical care in western countries, and account for most antibiotic prescriptions. While a confirmed and fast ARTI diagnosis is key for antibiotic prescribing, empiric antimicrobial treatment remains common, because viral symptoms are often clinically similar and difficult to distinguish from those caused by bacteria. As a result, inappropriate antibiotic prescriptions are high and in certain settings likely higher than the commonly estimated 30%. The QIAstat Respiratory Panel® assay (QIAstat RP) is a multiplexed in vitro diagnostics test for the rapid simultaneous detection of 21 pathogens directly from respiratory samples, including human mastadenovirus A-G, primate bocaparvovirus 1+2, human coronavirus (HKU1, NL63, OC43, 229E), human metapneumovirus A/B, rhinovirus/enterovirus, influenza A virus (no subtype, subtype H1, H1N1/2009, H3), influenza B virus, human respirovirus 1+3, human orthorubulavirus 2+4, human orthopneumovirus, Bordetella pertussis, Chlamydia pneumoniae, Mycoplasma pneumoniae and Legionella pneumophila. We describe the first multicenter study of 445 respiratory samples, collected through the 2016–2017 and 2018 respiratory seasons, with performance compared against BioFire FilmArray RP v1.7 and discrepancy testing by Seegene Allplex RP. The QIAstat RP demonstrated a positive percentage of agreement of 98.0% (95% CI: 96.0–99.1%) and a negative percentage agreement of 99.8% (95% CI: 99.6–99.9%). With use of this comprehensive and rapid test, improved patient outcomes and antimicrobial stewardship may potentially be achieved.
Collapse
Affiliation(s)
- Marijo Parčina
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Uffe Vest Schneider
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- * E-mail:
| | - Benoit Visseaux
- IAME, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, AP-HP, Laboratoire de Virologie, Hôpital Bichat, AP-HP, Paris, France
| | - Robert Jozić
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Irene Hannet
- STAT-Dx Life (Qiagen) Parc Científic, Barcelona, Spain
| | - Jan Gorm Lisby
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
33
|
Langeder J, Grienke U, Chen Y, Kirchmair J, Schmidtke M, Rollinger JM. Natural products against acute respiratory infections: Strategies and lessons learned. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112298. [PMID: 31610260 DOI: 10.1016/j.jep.2019.112298] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE A wide variety of traditional herbal remedies have been used throughout history for the treatment of symptoms related to acute respiratory infections (ARIs). AIM OF THE REVIEW The present work provides a timely overview of natural products affecting the most common pathogens involved in ARIs, in particular influenza viruses and rhinoviruses as well as bacteria involved in co-infections, their molecular targets, their role in drug discovery, and the current portfolio of available naturally derived anti-ARI drugs. MATERIALS AND METHODS Literature of the last ten years was evaluated for natural products active against influenza viruses and rhinoviruses. The collected bioactive agents were further investigated for reported activities against ARI-relevant bacteria, and analysed for the chemical space they cover in relation to currently known natural products and approved drugs. RESULTS An overview of (i) natural compounds active in target-based and/or phenotypic assays relevant to ARIs, (ii) extracts, and (iii) in vivo data are provided, offering not only a starting point for further in-depth phytochemical and antimicrobial studies, but also revealing insights into the most relevant anti-ARI scaffolds and compound classes. Investigations of the chemical space of bioactive natural products based on principal component analysis show that many of these compounds are drug-like. However, some bioactive natural products are substantially larger and have more polar groups than most approved drugs. A workflow with various strategies for the discovery of novel antiviral agents is suggested, thereby evaluating the merit of in silico techniques, the use of complementary assays, and the relevance of ethnopharmacological knowledge on the exploration of the therapeutic potential of natural products. CONCLUSIONS The longstanding ethnopharmacological tradition of natural remedies against ARIs highlights their therapeutic impact and remains a highly valuable selection criterion for natural materials to be investigated in the search for novel anti-ARI acting concepts. We observe a tendency towards assaying for broad-spectrum antivirals and antibacterials mainly discovered in interdisciplinary academic settings, and ascertain a clear demand for more translational studies to strengthen efforts for the development of effective and safe therapeutic agents for patients suffering from ARIs.
Collapse
Affiliation(s)
- Julia Langeder
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Ulrike Grienke
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria.
| | - Ya Chen
- University of Hamburg, Center for Bioinformatics (ZBH), Bundesstraße 43, 22763, Hamburg, Germany
| | - Johannes Kirchmair
- Department of Chemistry, University of Bergen, N-5020, Bergen, Norway; Computational Biology Unit (CBU), University of Bergen, N-5020, Bergen, Norway
| | - Michaela Schmidtke
- Section of Experimental Virology, Department of Medical Microbiology, Jena University Hospital, Hans-Knöll-Straße 2, Jena, 07745, Germany
| | - Judith M Rollinger
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| |
Collapse
|
34
|
Sarkar S, Heise MT. Mouse Models as Resources for Studying Infectious Diseases. Clin Ther 2019; 41:1912-1922. [PMID: 31540729 PMCID: PMC7112552 DOI: 10.1016/j.clinthera.2019.08.010] [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: 07/23/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Mouse models are important tools both for studying the pathogenesis of infectious diseases and for the preclinical evaluation of vaccines and therapies against a wide variety of human pathogens. The use of genetically defined inbred mouse strains, humanized mice, and gene knockout mice has allowed the research community to explore how pathogens cause disease, define the role of specific host genes in either controlling or promoting disease, and identify potential targets for the prevention or treatment of a wide range of infectious agents. This review discusses several of the most commonly used mouse model systems, as well as new resources such as the Collaborative Cross as models for studying infectious diseases.
Collapse
Affiliation(s)
- Sanjay Sarkar
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark T Heise
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. IMMUNITY & AGEING 2019; 16:25. [PMID: 31528180 PMCID: PMC6743147 DOI: 10.1186/s12979-019-0164-9] [Citation(s) in RCA: 319] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
The age-related dysregulation and decline of the immune system-collectively termed "immunosenescence"-has been generally associated with an increased susceptibility to infectious pathogens and poor vaccine responses in older adults. While numerous studies have reported on the clinical outcomes of infected or vaccinated individuals, our understanding of the mechanisms governing the onset of immunosenescence and its effects on adaptive immunity remains incomplete. Age-dependent differences in T and B lymphocyte populations and functions have been well-defined, yet studies that demonstrate direct associations between immune cell function and clinical outcomes in older individuals are lacking. Despite these knowledge gaps, research has progressed in the development of vaccine and adjuvant formulations tailored for older adults in order to boost protective immunity and overcome immunosenescence. In this review, we will discuss the development of vaccines for older adults in light of our current understanding-or lack thereof-of the aging immune system. We highlight the functional changes that are known to occur in the adaptive immune system with age, followed by a discussion of current, clinically relevant pathogens that disproportionately affect older adults and are the central focus of vaccine research efforts for the aging population. We conclude with an outlook on personalized vaccine development for older adults and areas in need of further study in order to improve our fundamental understanding of adaptive immunosenescence.
Collapse
Affiliation(s)
- Stephen N Crooke
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
36
|
Van Damme P, Coster ID, Bandyopadhyay AS, Suykens L, Rudelsheim P, Neels P, Oberste MS, Weldon WC, Clemens R, Revets H. Poliopolis: pushing boundaries of scientific innovations for disease eradication. Future Microbiol 2019; 14:1321-1330. [PMID: 31482728 DOI: 10.2217/fmb-2019-0196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Although global polio eradication is within reach, sustained eradication of all polioviruses requires cessation of oral poliovirus vaccine use to mitigate against vaccine-derived poliovirus circulation and vaccine-associated paralytic poliomyelitis. The first step in this direction was the WHO-recommended global withdrawal of live attenuated type 2 Sabin poliovirus from routine immunisation in May 2016, with future use restricted to outbreak response, and handling controlled by strict containment provisions (GAPIII). This creates unique challenges for development and testing of novel type 2 poliovirus vaccines. We describe the creation of a novel purpose-built containment facility, Poliopolis, to study new monovalent OPV2 vaccine candidates in healthy adult volunteers, which may be a model for future endeavors in vaccine development for emergency use.
Collapse
Affiliation(s)
- Pierre Van Damme
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| | - Ilse De Coster
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| | | | - Leen Suykens
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| | | | | | | | | | - Ralf Clemens
- Global Research in Infectious Diseases (GRID), Rio de Janeiro, Brazil
| | - Hilde Revets
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Belgium
| |
Collapse
|
37
|
Impact of Rhinovirus Infections in Children. Viruses 2019; 11:v11060521. [PMID: 31195744 PMCID: PMC6632063 DOI: 10.3390/v11060521] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/22/2019] [Accepted: 06/04/2019] [Indexed: 12/16/2022] Open
Abstract
Rhinovirus (RV) is an RNA virus that causes more than 50% of upper respiratory tract infections in humans worldwide. Together with Respiratory Syncytial Virus, RV is one of the leading causes of viral bronchiolitis in infants and the most common virus associated with wheezing in children aged between one and two years. Because of its tremendous genetic diversity (>150 serotypes), the recurrence of RV infections each year is quite typical. Furthermore, because of its broad clinical spectrum, the clinical variability as well as the pathogenesis of RV infection are nowadays the subjects of an in-depth examination and have been the subject of several studies in the literature. In fact, the virus is responsible for direct cell cytotoxicity in only a small way, and it is now clearer than ever that it may act indirectly by triggering the release of active mediators by structural and inflammatory airway cells, causing the onset and/or the acute exacerbation of asthmatic events in predisposed children. In the present review, we aim to summarize the RV infection's epidemiology, pathogenetic hypotheses, and available treatment options as well as its correlation with respiratory morbidity and mortality in the pediatric population.
Collapse
|
38
|
Osowicki J, Azzopardi KI, Baker C, Waddington CS, Pandey M, Schuster T, Grobler A, Cheng AC, Pollard AJ, McCarthy JS, Good MF, Walker MJ, Dale JB, Batzloff MR, Carapetis JR, Smeesters PR, Steer AC. Controlled human infection for vaccination against Streptococcus pyogenes (CHIVAS): Establishing a group A Streptococcus pharyngitis human infection study. Vaccine 2019; 37:3485-3494. [PMID: 31101422 DOI: 10.1016/j.vaccine.2019.03.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 12/17/2022]
Abstract
Group A Streptococcus (GAS) is a highly-adapted and human-restricted pathogen responsible for a high global burden of disease across a diverse clinical spectrum. Vaccine development has been impeded by scientific, regulatory, and commercial obstacles. Human infection studies (HIS) are increasingly contributing to drug, diagnostics, and vaccine development, reducing uncertainty at early stages, especially for pathogens with animal models that incompletely reproduce key elements of human disease. We review the small number of historical GAS HIS and present the study protocol for a dose-ranging inpatient study in healthy adults. The primary objective of the study is to establish a new GAS pharyngitis HIS with an attack rate of at least 60% as a safe and reliable platform for vaccine evaluation and pathogenesis research. According to an adaptive dose-ranging study design, emm75 GAS doses manufactured in keeping with principles of Good Manufacturing Practice will be directly applied by swab to the pharynx of carefully screened healthy adult volunteers at low risk of severe complicated GAS disease. Participants will remain as closely monitored inpatients for up to six days, observed for development of the primary outcome of acute symptomatic pharyngitis, as defined by clinical and microbiological criteria. All participants will be treated with antibiotics and followed as outpatients for six months. An intensive sampling schedule will facilitate extensive studies of host and organism dynamics during experimental pharyngitis. Ethics approval has been obtained and the study has been registered at ClinicalTrials.gov (NCT03361163).
Collapse
Affiliation(s)
- Joshua Osowicki
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Victoria, Australia; Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital Melbourne, Victoria, Australia.
| | - Kristy I Azzopardi
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ciara Baker
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Claire S Waddington
- Telethon Kids Institute, University of Western Australia and Perth Children's Hospital, Perth, Australia; Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Manisha Pandey
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Tibor Schuster
- Clinical Epidemiology and Biostatistics Unit, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia; Department of Family Medicine, McGill University, Montreal, Quebec, Canada
| | - Anneke Grobler
- Department of Paediatrics, University of Melbourne, Victoria, Australia; Clinical Epidemiology and Biostatistics Unit, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia
| | - Allen C Cheng
- Infection Prevention and Healthcare Epidemiology Unit, The Alfred Hospital, Melbourne, Victoria, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom; National Institute for Health Research, Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Medicine, University of Queensland, Brisbane, Australia
| | - Michael F Good
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - James B Dale
- University of Tennessee Health Science Center, Department of Medicine, Memphis, TN, USA
| | - Michael R Batzloff
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Jonathan R Carapetis
- Telethon Kids Institute, University of Western Australia and Perth Children's Hospital, Perth, Australia
| | - Pierre R Smeesters
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Victoria, Australia; Paediatric Department, Academic Children Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium; Molecular Bacteriology Laboratory, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrew C Steer
- Tropical Diseases, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Victoria, Australia; Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital Melbourne, Victoria, Australia
| |
Collapse
|
39
|
Girkin J, Maltby S, Singanayagam A, Bartlett N, Mallia P. In vivo experimental models of infection and disease. RHINOVIRUS INFECTIONS 2019. [PMCID: PMC7149593 DOI: 10.1016/b978-0-12-816417-4.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human and animal models continue to play a crucial role in research to understand host immunity to rhinovirus (RV) and identify disease mechanisms. Human models have provided direct evidence that RV infection is capable of exacerbating chronic respiratory diseases and identified immunological processes that correlate with clinical disease outcomes. Mice are the most commonly used nonhuman experimental RV infection model. Although semipermissive, under defined experimental conditions sufficient replication occurs to induce host immune responses that recapitulate immunity and disease during human infection. The capacity to use genetically modified mouse strains and drug interventions has shown the mouse model to be an invaluable research tool defining causal relationships between host immunity and disease and supporting development of new treatments. Used in combination the insights achieved from human and animal experimental infection models provide complementary insights into RV biology and yield novel therapeutic options to reduce the burden of RV-induced disease.
Collapse
|
40
|
Morgan DJ, Casulli J, Chew C, Connolly E, Lui S, Brand OJ, Rahman R, Jagger C, Hussell T. Innate Immune Cell Suppression and the Link With Secondary Lung Bacterial Pneumonia. Front Immunol 2018; 9:2943. [PMID: 30619303 PMCID: PMC6302086 DOI: 10.3389/fimmu.2018.02943] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Secondary infections arise as a consequence of previous or concurrent conditions and occur in the community or in the hospital setting. The events allowing secondary infections to gain a foothold have been studied for many years and include poor nutrition, anxiety, mental health issues, underlying chronic diseases, resolution of acute inflammation, primary immune deficiencies, and immune suppression by infection or medication. Children, the elderly and the ill are particularly susceptible. This review is concerned with secondary bacterial infections of the lung that occur following viral infection. Using influenza virus infection as an example, with comparisons to rhinovirus and respiratory syncytial virus infection, we will update and review defective bacterial innate immunity and also highlight areas for potential new investigation. It is currently estimated that one in 16 National Health Service (NHS) hospital patients develop an infection, the most common being pneumonia, lower respiratory tract infections, urinary tract infections and infection of surgical sites. The continued drive to understand the mechanisms of why secondary infections arise is therefore of key importance.
Collapse
Affiliation(s)
- David J Morgan
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Joshua Casulli
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christine Chew
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Emma Connolly
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Sylvia Lui
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Oliver J Brand
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Rizwana Rahman
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christopher Jagger
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
41
|
Wang Y, Dong T, Qi G, Qu L, Liang W, Qi B, Zhang Z, Shang L, Gao H, Du X, Lu B, Guo Y, Liu Z, Yu H, Cui Q, Wang X, Li Y, Guo W, Qu Z. Prevalence of Common Respiratory Viral Infections and Identification of Adenovirus in Hospitalized Adults in Harbin, China 2014 to 2017. Front Microbiol 2018; 9:2919. [PMID: 30542337 PMCID: PMC6277751 DOI: 10.3389/fmicb.2018.02919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/14/2018] [Indexed: 01/05/2023] Open
Abstract
Background: Respiratory infections pose a great challenge in global health, and the prevalence of viral infection in adult patients has been poorly understood in northeast China. Harbin is one of the major cities in northeast China, and more than half of any given year in Harbin is occupied by winter. To reveal the viral etiology and seasonality in adult patients from Harbin, a 4-year consecutive survey was conducted in Harbin, China. Methods: From January 2014 to December 2017, specimens were obtained from adult patients admitted to the Second Affiliated Hospital of Harbin Medical University with lower respiratory tract infections. Sputum samples were examined by direct immunofluorescence assays to detect seven common respiratory viruses, including influenza virus (type A and B), parainfluenza virus (type 1 to 3), respiratory syncytial virus and adenovirus. Adenovirus positive samples were seeded onto A549 cells to isolate viral strains. Phylogenetic analysis was conducted on the highly variable region of adenoviral hexon gene. Results: A total of 1,300 hospitalized adult patients with lower respiratory tract infections were enrolled, in which 189 patients (14.5%) were detected as having at least one viral infection. The co-infection rate in this study was 25.9% (49/189). The dominant viral pathogen from 2014 to 2017 was parainfluenza virus, with a detection rate of 7.2%, followed by influenza virus, respiratory syncytial virus and adenovirus. Based on the climate seasons determined by daily average temperature, the highest overall viral detection rate was detected in spring (22.0%, 52/236), followed by winter (13.4%, 109/813), autumn (11.4%, 13/114) and summer (10.9%, 15/137). Adenovirus type 3 strains with slight variations were isolated from positive cases, which were closely related to the GB strain from the United States, as well as the Harbin04B strain isolated locally. Conclusion: This study demonstrated that common respiratory viruses were partially responsible for hospitalized lower respiratory tract infections in adult patients from Harbin, China, with parainfluenza virus as the dominant viral pathogen. Climate seasons could be rational indicators for the seasonality analysis of airborne viral infections. Future surveillance on viral mutations would be necessary to reveal the evolutionary history of respiratory viruses.
Collapse
Affiliation(s)
- Yingchen Wang
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Tuo Dong
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Guiyun Qi
- Department of Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lixin Qu
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Wei Liang
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Binbin Qi
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Zhe Zhang
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Lei Shang
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Hong Gao
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Xiqiao Du
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Bing Lu
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Yan Guo
- Department of Ear Nose Throat, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenwei Liu
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Huisong Yu
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Qi Cui
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Xiaocen Wang
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Ye Li
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China
| | - Weiyuan Guo
- Department of Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhangyi Qu
- Department of Microbiology, Public Health College, Harbin Medical University, Harbin, China.,Department of Natural Focus Disease Control, Institute of Environment-Associated Disease, Sino-Russia Joint Medical Research Center, Harbin Medical University, Harbin, China
| |
Collapse
|