1
|
Munro APS, Drysdale SB, Cathie K, Flamein F, Knuf M, Collins AM, Hill HC, Kaiser F, Cohen R, Pinquier D, Vassilouthis NC, Carreno M, Moreau C, Bourron P, Marcelon L, Mari K, Roberts M, Tissières P, Royal S, Faust SN, HARMONIE Study Group. 180-day efficacy of nirsevimab against hospitalisation for respiratory syncytial virus lower respiratory tract infections in infants (HARMONIE): a randomised, controlled, phase 3b trial. THE LANCET. CHILD & ADOLESCENT HEALTH 2025; 9:404-412. [PMID: 40379431 DOI: 10.1016/s2352-4642(25)00102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection and hospitalisations in infants worldwide. The primary analyses of HARMONIE showed that nirsevimab reduced infant hospitalisations due to RSV-associated lower respiratory tract infection through the RSV season. This analysis aims to evaluate nirsevimab's efficacy at 180 days after dosing, a period exceeding the typical 5-month RSV season. METHODS HARMONIE is an ongoing, open-label, parallel arm, randomised, controlled, phase 3b study conducted in France, Germany, and the UK. Infants aged 12 months or younger, born at a gestational age of at least 29 weeks, were randomly assigned (1:1) to receive either a single intramuscular dose of nirsevimab (50 mg for children <5 kg or 100 mg for children ≥5 kg) or standard care (without RSV prophylaxis) before or during their first RSV season. Randomisation was electronically done, stratified by country and age-group. The primary efficacy endpoint for this analysis was the incidence of hospitalisations due to RSV-associated lower respiratory tract infection up to 180 days after nirsevimab administration or randomisation in all randomised participants. Safety up to 365 days following nirsevimab administration was also assessed. This trial is ongoing and registered with ClinicalTrials.gov, number NCT05437510. FINDINGS Between Aug 8, 2022, and Feb 28, 2023, 8057 infants were randomly assigned to either the nirsevimab group (n=4038) or the standard care group (n=4019). The median age at randomisation was 4·00 months (IQR 1·0-7·0; range 0·0-12·0, and 4195 (52·1%) were male and 3862 (47·9%) were female. Up to 180 days, 12 (0·3%) of 4038 infants in the nirsevimab group and 68 (1·7%) of 4019 infants in the standard care group had been hospitalised for RSV-associated lower respiratory tract infection, corresponding to a nirsevimab efficacy of 82·7% (95% CI 67·8-91·5; p<0·0001). Most participants experienced grade 1 (2759 [68·7%] of 4016 in the nirsevimab group; 2696 [67·1%] of 4018 in the standard care group) or grade 2 (1447 [36·0%] of 4016 in the nirsevimab group; 1436 [35·7%] of 4018 in the standard care group) treatment-emergent adverse events, and no apparent safety concerns were raised up to 365 days after dosing. INTERPRETATION Nirsevimab offers consistent and sustained protection against hospitalisation due to RSV-associated lower respiratory tract infection for at least 6 months. This finding provides global health systems greater flexibility when implementing nirsevimab, providing substantial benefit in the ongoing effort to reduce the burden of infant RSV and the potential wider public health value. FUNDING Sanofi and AstraZeneca.
Collapse
Affiliation(s)
- Alasdair P S Munro
- National Institute for Health and Care Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Simon B Drysdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; The NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Katrina Cathie
- National Institute for Health and Care Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Florence Flamein
- Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille, France; French Clinical Research Infrastructure Network-PEDSTART, Tours, France
| | - Markus Knuf
- Children's Hospital, Worms, Germany; Pediatric Infectious Diseases, University Medicine, Mainz, Germany
| | - Andrea M Collins
- Liverpool Vaccine Group, Liverpool School of Tropical Medicine, Liverpool, UK; Liverpool University Hospitals Foundation NHS Trust, Liverpool, UK
| | - Helen C Hill
- Liverpool Vaccine Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Friedrich Kaiser
- Gemeinschaftspraxis für Kinder und Jugendmedizin, Hamburg, Germany
| | - Robert Cohen
- Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil, France
| | - Didier Pinquier
- Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen, France
| | | | | | | | | | | | | | | | - Pierre Tissières
- IHU-PROMETHEUS Comprehensive Sepsis Center and Pediatric Intensive Care, Neonatal Medicine, and Pediatric Emergency Department, AP-HP Paris Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre, France; Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette, France
| | - Simon Royal
- University of Nottingham Health Service, University of Nottingham, Nottingham, UK
| | - Saul N Faust
- National Institute for Health and Care Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK.
| | | |
Collapse
Collaborators
France, Jacques Brouard, Andre Leke, Veronique Brevaut-Malaty, Chloe Epiard, Florence Flamein, Ralph Epaud, David Somerville, Franck Thollot, Phu My Tran Dinh, Mathie Lorrot, Andreas Werner, Rachel Froget, Hasinirina Razafimahefa, Michael Fayon, Camille Brehin, Pascal Boileau, Kim-An Nguyen, Cecile Guiheneuf, Claire Lefevre, Claire Sarreau, Fabienne Kochert, Frederic Huet, Elisa Seror, Fabienne Cahn-Sellem, Nadege Delavie, Christophe Batard, François Corrard, Morched Zouari, Nathalie Gelbert-Baudino, Diane Carriere, Zoha Maakaroun-Vermesse, Jean Gaschignard, Stephane Rioualen, Frederic Dugelay, Alain Wollner, Kai Kassmann, Didier Pinquier, Jean-Francois Delobbe, Robert Cohen, Anne Cheve, Anne Bourlet, Christele Gras-Le Guen, Benedicte Nold, Bergengere Kireche, Anne-Sylvestre Michot-Cottias, Francine Lecaillier, Myrna Achkar, Lisa Giovannini-Chami, Aimen Bsila, Laura Vivalda, Florence Goehringer, Amelie Poidvin, Christine Magendie, Emilie Georget, Blandine Desse, Vincent Gajdos, Didier Pinquier, Eric Jeziorski, Sanaa Naji, Christian Petit, Mallorie Mondenx, Emmanuelle Rondeleux, Aurelie Morand, Cecile Kerdudo-Veau, Blandine Prevost, Alexandra David, Gaelle Cornen, Christine Devulder, Sven Wellmann, Cordula Koerner-Rettberg, Otto Laub, Hartmut Scheele, Manfred Praun, Markus Knuf, Martin Wetzke, Helmut Pabel, Andreas Petri, Ulrich Thome, Falko Panzer, Hans Fuchs, Jana-Katharina Dieks, Martin Oliver Bauer, Jost Richter, Katrin Biebach, Frank Radowsky, Matthias Donner, Katja Denneberg, Jurgen Funck, Soeren Westerholt, Michael Horn, Malte Cremer, Wolfgang Kamin, Ulf Schulze-Sturm, Nikolaos Konstantopoulos, Brigitte Wilmsmeyer, Thomas Voelkl, Martina von Poblotzki, Andreas Mueller, Egbert Leonhardt, Siegfried Simmet, Eivy Franke-Beckmann, Stefan Eber, Thorsten Froehlich, Matthias Gorenflo, Eckard Hamelmann, Tobias Borchers, Ralph Koellges, Frank Jochum, Florian Brinkert, Monika Gappa, Christiane Lex, Sonja Behrendt, Adriana Haus, Eva Hahn, Sebastian Wirth, Ralph Maier, Sebastian Horn, Gergely Sarkoezy, Katja Weiss, Lutz Hempel, Friedrich Ebinger, Franziska Schaaff, Tilman Humpl, Torsten Ott, Louisa van den Boom, Dominik Schneider, Stephan Gehring, Ludwig Stapenhorst, Donald Wurm, Stefan Debler, Viola Gerstmann, Ralph Melchior, Joel Iffland, Norbert Teig, Tina-Maria Weichert, Thomas Glatzel, Eva Galiza, Elizabeth Whittaker, Nick Wooding, Sunil Bhimsaria, Patrick Moore, Nick Thomas, Sanjay Salgia, Anu Goenka, Timothy Johnson, Mildred Iro, Shye-Wei Wong, Arshid Murad, Samuel Oddie, Kelechi Ugonna, Clare Murray, Balaji Suryanarayanan, Nicola Millen, Sian Ludman, Nafeesa Arshad, Aung Soe, John Jackson, Manu Vatish, Dominic Smith, Paul Clarke, Chidambara Harikumar, Nicola Lester, Srinivas Bandi, Simon Royal, Athanasios Konstantinidis, Daniel Hawcutt, Lawrence Barnes, Rebecca Mann, Carrie Heal, Dinakaran Jayachandran, Amardeep Heer, Dinakaran Jayachandran, Charusheela Bhatia, Paul Fleming, Julian Forton, Ahmet Fuat, Basheer Peer-Mohamed, Victoria Robinson, Marieke Emonts, Nicholas Jacobsen, Samuel Davies, Girish Gowda, Camelia Vaina, Katrina Cathie, Otilia Popescu, James Kennard, Haji Khan, John Chapman, Krishna Jada, Jose-Ramon Fernandez-Alvarez, Prakash Kamath, Ian Binnian, Sarah Rayne, Clara Thompson, Binu Anand, Clare Hollingsworth, Timothy Scorrer, Vennila Ponnusamy, Christina Oliver, Christopher Bedford, Nilanjana Ray, Seethwakage Seram, Carolyn Paul, Damien McNally, William Priestman, William White, Stuart Mackay-Thomas, Claire Chalmers-Watson, Nilesh Agrawal, Atul Gupta, John Ryan, Alan Cade, Matthew Gaw, Claire Hombersley, Jonathan Cohen, Sweta Jain, Thomas Morgan, Clare Webster, Tamsin Simpkins, Mo Roshan, David Lewis, Katharine McDevitt, Mark Tighe, Charles Deakin, Vimal Vasu, Bushra Abdul Malik, Michael Yanney, James Perry, Ruth Cowie, Helen Purves, Bindu Radha, Lucinda Winckworth, Mark Danielsen, Rosaline Garr, Sanjay Rawal, Pete Wilson, Zelda Cheng, Adejumoke Awoseyila, Sebastian Gray, Lucy Martin, Laurence Cribbin, Rebecca Brown, Richa Gupta, Kevin Pender, Graham Roberts, Richard Burkimsher,
Collapse
|
2
|
Karthigeyan KP, Connors M, Binuya CR, Gross M, Fuller AS, Crooks CM, Wang HY, Sponholtz MR, Byrne PO, Herbek S, Andy C, Gerber LM, Campbell JD, Williams CA, Mitchell E, van der Maas L, Miller I, Yu D, Bottomley MJ, McLellan JS, Permar SR. A human cytomegalovirus prefusion-like glycoprotein B subunit vaccine elicits humoral immunity similar to that of postfusion gB in mice. J Virol 2025:e0217824. [PMID: 40338082 DOI: 10.1128/jvi.02178-24] [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: 12/09/2024] [Accepted: 04/06/2025] [Indexed: 05/09/2025] Open
Abstract
Human cytomegalovirus (HCMV) is the leading infectious cause of birth defects. Despite the global disease burden, there is no Food and Drug Administration (FDA)-approved HCMV vaccine. The most efficacious HCMV vaccine candidates to date have used glycoprotein B (gB), a class III viral fusion protein, in its postfusion form. While some viral fusion proteins have been shown to elicit stronger neutralizing responses in their prefusion conformation, HCMV prefusion-like and postfusion gB were recently shown to elicit antibodies with similar fibroblast neutralization titers in mice. We aimed to define and compare the specificity and functionality of plasma IgG elicited by distinct prefusion-like and postfusion gB constructs. Prefusion-like and postfusion gB elicited comparable IgG responses that predominantly mapped to the AD-5 antigenic domain known to elicit neutralizing antibodies. Interestingly, postfusion gB elicited significantly higher plasma IgG binding to cell-associated gB and antibody-dependent cellular phagocytosis than that of prefusion-like gB. The vaccines elicited comparable neutralization titers of heterologous HCMV strain AD169r in fibroblasts; however, neither elicited neutralizing titers against the vaccine-matched strain Towne in fibroblasts. Our data indicate that gB in this prefusion-like conformation elicits similar specificity and functional humoral immunity to that of postfusion gB, unlike certain class I viral fusion proteins that have been used as vaccine antigens. These findings deepen our understanding of the immune response elicited by class III fusion proteins and may inform further design and testing of conformationally dependent herpesvirus glycoprotein vaccine candidates.IMPORTANCEVaccines against human cytomegalovirus (HCMV) still remain elusive in spite of the high disease burden of the virus, especially in pre-term infants and immunocompromised individuals. While vaccine efforts have focused on vaccine-induced antibodies to neutralize the virus, studies have increasingly shown the importance of other antibody functions in protection against cytomegalovirus (CMV) transmission. In this study, we comprehensively evaluated immune responses elicited by the prefusion state of an important HCMV protein called glycoprotein B (gB) in mice. Our results indicate that prefusion gB elicits immune responses similar to that of postfusion gB in mice and reveals areas for further redesign and testing for prefusion vaccine antigens against CMV and other herpesviruses, which could help in furthering vaccine development against HCMV.
Collapse
Affiliation(s)
| | - Megan Connors
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Christian R Binuya
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Mackensie Gross
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Adelaide S Fuller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Chelsea M Crooks
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Madeline R Sponholtz
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Patrick O Byrne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - Linda M Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - John D Campbell
- Dynavax Technologies Corporation, Emeryville, California, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Elizabeth Mitchell
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Lara van der Maas
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Itzayana Miller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Dong Yu
- Dynavax Technologies Corporation, Emeryville, California, USA
| | | | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
3
|
Wu PP, Ding FR. Administration of Nirsevimab for RSV Prophylaxis in Infants: A Comprehensive Review. Vaccines (Basel) 2025; 13:470. [PMID: 40432081 PMCID: PMC12115584 DOI: 10.3390/vaccines13050470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Respiratory syncytial virus (RSV) is the primary etiological agent responsible for lower respiratory tract infections (LRTIs) and hospitalizations among infants. Nirsevimab, a novel monoclonal antibody (mAb), offers sustained protection against RSV for a minimum of 5 months in neonates and young children. Extensive clinical trials and real-world evidence have demonstrated that nirsevimab significantly mitigates the incidence and severity of RSV infections in infants, while exhibiting favorable safety profiles and cost-effectiveness. Regulatory authorities in multiple countries have approved nirsevimab, and its implementation is progressively expanding across various healthcare settings. However, several critical issues require further attention. Specifically, a more in-depth investigation into the long-term efficacy and benefits of nirsevimab across diverse populations, particularly neonates, is essential. Additionally, accelerating the introduction and administration of nirsevimab in developing countries remains imperative. Thus, this review comprehensively summarizes the administration of nirsevimab in infants to facilitate its broader application.
Collapse
Affiliation(s)
- Pan-Pan Wu
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, China;
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin 300100, China
| | - Fang-Rui Ding
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin 300100, China;
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin 300100, China
| |
Collapse
|
4
|
Wang X, Kong L, Liu X, Wu P, Zhang L, Ding F. Effectiveness of nirsevimab immunization against RSV infection in preterm infants: a systematic review and meta-analysis. Front Immunol 2025; 16:1581970. [PMID: 40313952 PMCID: PMC12043638 DOI: 10.3389/fimmu.2025.1581970] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Background Respiratory Syncytial Virus (RSV) is one of the primary pathogen responsible for severe lower respiratory tract infections in preterm infants, placing a significant burden on patients, their families, and society. Nirsevimab, a recently developed RSV monoclonal antibody, has demonstrated promising efficacy in this population according to preliminary studies. However, there remains a need for comprehensive systematic reviews and meta-analyses to evaluate the effectiveness of nirsevimab in preventing RSV-related lower respiratory tract infections in preterm infants. Methods A search of the PubMed and EMBASE databases was conducted to identify randomized controlled trials (RCTs) and observational studies assessing the prevention of RSV infection in preterm infants using nirsevimab. Relevant data were extracted and subjected to meta-analysis. Results Five studies involving a total of 7,347 preterm infants (3,987 in the nirsevimab group and 3,360 in the control group) were included. The meta-analysis revealed that nirsevimab significantly reduced the incidence of medically attended RSV-associated lower respiratory tract infections (OR = 0.25; 95% CI: 0.15, 0.40; P < 0.0001) and hospitalizations due to RSV-associated lower respiratory tract infections (OR = 0.27; 95% CI: 0.19, 0.38; P < 0.0001). Conclusion Nirsevimab significantly decreases the risk of RSV-related infection in preterm infants and represents a valuable intervention for RSV prevention in this vulnerable population. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42025629937.
Collapse
Affiliation(s)
- Xiaopeng Wang
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin, China
| | - Li Kong
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin, China
| | - Xueou Liu
- Research Institute of Obstetrics and Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Panpan Wu
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin, China
| | - Lulu Zhang
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin, China
| | - Fangrui Ding
- Department of Neonatology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Neonatology, Nankai University Maternity Hospital, Tianjin, China
| |
Collapse
|
5
|
Plock N, Sachs JR, Zang X, Lommerse J, Vora KA, Lee AW, Cheung SYA, Maas BM. Efficacy of monoclonal antibodies and maternal vaccination for prophylaxis of respiratory syncytial virus disease. COMMUNICATIONS MEDICINE 2025; 5:119. [PMID: 40240559 PMCID: PMC12003833 DOI: 10.1038/s43856-025-00807-9] [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: 09/18/2023] [Accepted: 03/14/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a leading cause of respiratory tract infection in infants and young children. The level of serum neutralizing antibodies (SNAs) is often used as a measure of protection against respiratory syncytial virus (RSV) infection. METHODS A qualified, model-based, meta-analysis efficacy prediction framework was used to understand the maternal vaccination-induced fold-increase in SNA titers necessary to achieve, over several study observation periods and study populations, similar protection to that of the monoclonal antibody clesrovimab (MK-1654). RESULTS Simulations indicated that 3-month and 6-month efficacy comparable to that predicted for passive immunization (clesrovimab) would require a maternal vaccine to increase SNA titers by 30- and 60-fold, respectively, higher than observed increases reported to date. Efficacy of maternal vaccination was predicted (for vaccines similar to those with published data) to be substantially lower for preterm infants compared to full-term infants, and substantially less over 6 months than over 3 months. Efficacy of passive immunization was predicted to be similar or higher in preterm infants than full-term infants and was similar for 3- and 6-month observation periods. CONCLUSIONS Modeling can be used to reliably predict the efficacy of maternal vaccination for preventing RSV in infants. Passive immunization (e.g., with clesrovimab) is likely to provide more protection for preterm infants and for infants born outside the RSV season than that provided by current maternal vaccines. Maternal vaccination may provide partial protection from RSV disease to full-term infants born just prior to or during the RSV season.
Collapse
Affiliation(s)
| | | | | | - Jos Lommerse
- Certara, Radnor, PA, USA
- Nalma, Oss, The Netherlands
| | | | - Andrew W Lee
- Merck & Co., Inc., Rahway, NJ, USA
- Uniquity Bio, Malvern, PA, USA
| | | | | |
Collapse
|
6
|
Liu X, Balligand T, Le Gall C, Ploegh HL. A monoclonal anti-hemagglutinin stem antibody modified with zanamivir protects against both influenza A and B viruses. Proc Natl Acad Sci U S A 2025; 122:e2424889122. [PMID: 40193611 PMCID: PMC12012527 DOI: 10.1073/pnas.2424889122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/01/2025] [Indexed: 04/09/2025] Open
Abstract
Influenza remains a significant public health threat. Both monoclonal antibodies and small-molecule inhibitors can target the influenza surface glycoproteins hemagglutinin (HA) or neuraminidase (NA) for prevention and treatment of influenza. Here, we combine the strengths of anti-influenza antibodies and small molecules by site-specific conjugation of the NA inhibitor zanamivir to MEDI8852, an HA-specific fully human monoclonal antibody. MEDI8852 targets the conserved stem region of HA and inhibits HA-mediated fusion of the viral and host cell membranes. Elimination of virus-infected cells involves Fcγ receptor-mediated effector functions. The efficacy of MEDI8852 is limited to influenza A viruses. Zanamivir, on the other hand, binds to the active site of NA in both influenza A and B viruses to inhibit NA activity and virus release. However, because of its small size, zanamivir has a short half-life and requires repeated dosing at high concentrations. We produced a MEDI8852-zanamivir antibody-drug conjugate (ADC) that engages Fc-mediated effector functions and benefits from neonatal Fc receptor (FcRn)-mediated recycling. The MEDI8852-zanamivir conjugate extends the circulatory half-life of zanamivir, targets both influenza HA and NA, and shows enhanced antibody-dependent cellular cytotoxicity (ADCC) compared to MEDI8852 alone. The MEDI8852-zanamivir conjugate protected mice from a lethal (10 × LD50) challenge with influenza A and B viruses at a dose similar to that required for broadly neutralizing anti-NA antibodies, with the added advantage of simultaneously targeting NA (influenza A and B) and HA (influenza A).
Collapse
Affiliation(s)
- Xin Liu
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Thomas Balligand
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Camille Le Gall
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Hidde L. Ploegh
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| |
Collapse
|
7
|
Thambi N, Phuah JY, Staupe RP, Tobias LM, Cao Y, McKelvey T, Railkar RA, Aliprantis AO, Arriola CS, Maas BM, Vora KA. Development of High-Titer Antidrug Antibodies in a Phase 1b/2a Infant Clesrovimab Trial Are Associated With RSV Exposure Beyond Day 150. J Infect Dis 2025; 231:e488-e496. [PMID: 39590882 PMCID: PMC11911791 DOI: 10.1093/infdis/jiae582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/27/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Clesrovimab is a human half-life-extended monoclonal antibody in phase 3 evaluation for the prevention of respiratory syncytial virus (RSV) disease in infants. Antidrug antibodies (ADA) were observed at late time points in a phase 1b/2a study where clesrovimab was well tolerated with an extended half-life of approximately 45 days. METHODS Serum samples at days 150, 365, and 545 postdose were assayed for ADA titers. Samples with high ADA titers were characterized for their binding specificity to the Fab or the YTE portion of clesrovimab. RSV serum neutralization (SNA) titers were also measured on ADA-positive and ADA-negative infants. Additionally, a D25 (site Ø) competitive enzyme-linked immunosorbent assay (ELISA) was performed on ADA-positive available samples to determine RSV exposure. Local surveillance data was used to ascertain RSV circulation during the trial. RESULTS High ADA titers were observed in a minority of infants at days 365 and 545 for all doses tested. Additionally, all high-titer ADA-positive infants had ADA directed towards the YTE epitope of clesrovimab. Moreover, these infants demonstrated robust RSV-SNA and had D25 competitive antibodies suggesting an RSV exposure after day 150, coinciding with the epidemiological data. CONCLUSIONS RSV exposure in infants beyond day 150 after dosing is associated with ADA development and high RSV-SNA titers with no impact on pharmacokinetics. CLINICAL TRIALS REGISTRATION NCT03524118.
Collapse
Affiliation(s)
| | | | | | - Lori M Tobias
- Department of Infectious Diseases and Vaccines
- Department of Pharmacokinetics Dynamics Metabolism Bioanalytics
| | - Yu Cao
- Department of Infectious Diseases and Vaccines
- Department of Pharmacokinetics Dynamics Metabolism Bioanalytics
| | - Troy McKelvey
- Department of Pharmacokinetics Dynamics Metabolism Bioanalytics
| | - Radha A Railkar
- Department of Infectious Diseases and Vaccines
- Department of Biostatistics and Research Decision Sciences
| | | | | | - Brian M Maas
- Department of Infectious Diseases and Vaccines
- Department of Quantitative Pharmacology & Pharmacometrics, Merck & Co., Inc., Rahway, New Jersey, USA
| | | |
Collapse
|
8
|
Madhi SA, Simões EAF, Acevedo A, Novoa Pizarro JM, Shepard JS, Railkar RA, Cao X, Maas BM, Zang X, Krick A, Roadcap B, Vora KA, Aliprantis AO, Lee AW, Sinha A. A Phase 1b/2a Trial of a Half-life Extended Respiratory Syncytial Virus Neutralizing Antibody, Clesrovimab, in Healthy Preterm and Full-term Infants. J Infect Dis 2025; 231:e478-e487. [PMID: 39601265 PMCID: PMC11911779 DOI: 10.1093/infdis/jiae581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Clesrovimab is an investigational monoclonal antibody with an extended half-life targeting site IV of the respiratory syncytial virus (RSV) fusion protein for the prevention of RSV disease in infants. METHODS In this phase 1b/2a, double-blind study, 183 healthy preterm and full-term infants 2 weeks to 8 months of age were randomized 4:1 within 5 panels (preterm 20, 50, 75, or 100 mg; full-term 100 mg) to receive 1 dose of clesrovimab or placebo. The objectives were to evaluate safety, pharmacokinetics, serum neutralizing antibodies (SNA), and antidrug antibodies (ADA). The incidence of RSV-associated end points (medically attended lower respiratory tract infection, hospitalization, and acute respiratory infection) were also evaluated through 150 days postdose. RESULTS The most common adverse event through day 14 was irritability; no treatment-related serious AEs were reported. Clesrovimab serum concentrations displayed a geometric mean apparent half-life of 44.9 days. Of participants receiving clesrovimab, 51 (36.7%) developed ADA with no apparent impact in pharmacokinetics. SNA titers increased in a dose-dependent manner at day 150. The incidences of RSV-associated end points were lower in infants treated with clesrovimab compared with placebo. CONCLUSIONS Clesrovimab was generally well tolerated and exhibited an extended half-life compared to typical IgG1 antibodies, supporting its ongoing development in late-stage trials. Clinical Trial Registration. NCT03524118.
Collapse
MESH Headings
- Humans
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Infections/drug therapy
- Double-Blind Method
- Male
- Female
- Antibodies, Neutralizing/blood
- Half-Life
- Infant
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Infant, Premature
- Infant, Newborn
- Antibodies, Viral/blood
- Respiratory Syncytial Virus, Human/immunology
- Antiviral Agents/pharmacokinetics
- Antiviral Agents/adverse effects
- Antiviral Agents/administration & dosage
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/administration & dosage
Collapse
Affiliation(s)
- Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Eric A F Simões
- Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
- Colorado School of Public Health, Aurora, Colorado, USA
| | | | | | | | | | - Xin Cao
- Merck & Co, Inc, Rahway, New Jersey, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Fly JH, Eiland LS, Stultz JS. Nirsevimab: Expansion of Respiratory Syncytial Virus Prevention Options in Neonates, Infants, and At-Risk Young Children. Ann Pharmacother 2025; 59:81-91. [PMID: 38654469 DOI: 10.1177/10600280241243357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
OBJECTIVE Review available data from clinical trials of nirsevimab for efficacy and safety in the setting of respiratory syncytial virus (RSV) prophylaxis in infants and children, while exploring nirsevimab's role in clinical practice and highlighting continuing questions. DATA SOURCES A literature search of PubMed was conducted utilizing the phrases "nirsevimab" and "medi8897." Additional references were identified through found references. Organizational guidelines, medication labeling, and regulatory organization presentations were utilized. STUDY SELECTION AND DATA EXTRACTION Relevant clinical trials investigating nirsevimab in infants and children were included as well as other references on pharmacology, pharmacokinetics, and pharmacoeconomics. DATA SYNTHESIS Nirsevimab, a once-a-season monoclonal antibody, demonstrated a 79.5% (95% CI, 65.9-87.7; P < 0.00001) lower incidence of RSV-associated medically attended lower respiratory tract infections (MA RSV-associated LRTI) and 77.3% (95% CI, 50.3-89.7; P = 0.0002) reduction in hospitalizations for RSV-associated MA-LRTI across 2 placebo-controlled studies. Nirsevimab demonstrated comparable safety to placebo with minor dermatologic reactions being the most common adverse event (0.9% vs 0.6%). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IN COMPARISON WITH EXISTING AGENTS Nirsevimab was approved by the US Food and Drug Administration, and recommended by the Advisory Committee on Immunization Practices and American Academy of Pediatrics for broad administration to infants entering their first RSV season and at risk patients during their second RSV season. Questions remain over efficacy in infants born < 29-week gestation and other economical considerations. CONCLUSIONS Nirsevimab demonstrated clinical efficacy in reducing RSV-associated MA-LRTI and RSV-associated hospitalizations in infants and was well tolerated.
Collapse
Affiliation(s)
- J Hunter Fly
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
- Le Bonheur Children's Hospital, Memphis, TN, USA
| | - Lea S Eiland
- Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jeremy S Stultz
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, The University of Tennessee Health Science Center, Nashville, TN, USA
| |
Collapse
|
10
|
Mao X, Hua X, Wu C, Ge X, Zhang J, Wu X, Kubiak RJ, Wählby Hamrén U, Villafana T, Christou G, Green J, Takas T, Jin Y. A phase I, randomized, placebo-controlled trial to evaluate the pharmacokinetics, safety, and tolerability of nirsevimab in healthy Chinese adults. Clin Transl Sci 2025; 18:e70095. [PMID: 39736101 DOI: 10.1111/cts.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/18/2024] [Accepted: 11/07/2024] [Indexed: 01/01/2025] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection (LRTI) in infants worldwide. Nirsevimab, an extended half-life monoclonal antibody against RSV, is approved in China for the prevention of RSV lower respiratory tract disease in infants; however, global nirsevimab trials did not enroll Chinese infants. To inform the investigation of nirsevimab for the prevention of RSV LRTI in Chinese infants, this Phase I, randomized, placebo-controlled trial evaluated the pharmacokinetics (PK) and safety of nirsevimab in healthy Chinese adults. Participants were randomized 3:1 to a single 300 mg intramuscular dose of nirsevimab or placebo and were followed through 150 days post-dose. Serum nirsevimab concentrations were measured and PK parameters of maximum serum concentration (Cmax), time to maximum concentration (tmax), and area under the concentration-time curve from time 0 to Day 150 (AUC0-150) were estimated. Treatment emergent adverse events (AEs), clinical laboratory data, and vital signs were evaluated. Overall, 24 participants were randomized to nirsevimab (n = 18) or placebo (n = 6). Nirsevimab geometric mean (coefficient of variation [%CV]) Cmax was 46.9 (21.7) μg/mL, median (range) tmax was 7.0 (4.9, 29.9) days, and geometric mean (%CV) AUC0-150 was 4210.6 (13.6) μg·day/mL. Treatment-emergent AEs (all Grade 1 or Grade 2 in severity) were reported in 5/18 (27.8%) nirsevimab recipients and 2/6 (33.3%) placebo recipients. No serious AEs, new onset chronic disease, or deaths were reported. Overall, safety and PK outcomes were consistent with those observed in healthy adults in the USA, with no new safety concerns.
Collapse
Affiliation(s)
- Xiaomeng Mao
- Clinical Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohan Hua
- Clinical Development, Respiratory & Immunology, R&D China, AstraZeneca, Shanghai, China
| | - Chengyi Wu
- Biometrics, R&D China, AstraZeneca, Shanghai, China
| | - Xiaoyun Ge
- Clinical Safety, R&D China, AstraZeneca, Shanghai, China
| | - Jie Zhang
- Development Operations, R&D China, AstraZeneca, Shanghai, China
| | - Xiaojie Wu
- Clinical Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Robert J Kubiak
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tonya Villafana
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Georgios Christou
- Patient Safety, Chief Medical Office, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Jannine Green
- Global Clinical Operations, R&D, AstraZeneca, Macclesfield, UK
| | - Therese Takas
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Yuwen Jin
- Clinical Pharmacology, R&D China, AstraZeneca, Shanghai, China
| |
Collapse
|
11
|
Ocana de Sentuary C, Testard C, Lagrée M, Leroy M, Gasnier L, Enes-Dias A, Leruste C, Diallo D, Génin M, Rakza T, Dubos F. Acceptance and safety of the RSV-preventive treatment of newborns with nirsevimab in the maternity department: a prospective longitudinal cohort study in France. EClinicalMedicine 2025; 79:102986. [PMID: 39726670 PMCID: PMC11669793 DOI: 10.1016/j.eclinm.2024.102986] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Background To evaluate the acceptance and safety of the treatment of newborns with nirsevimab (a long-acting monoclonal antibody designed to prevent respiratory syncytial virus infections) during the first season of implementation. Methods A longitudinal, prospective, single-centre cohort study was conducted from September 18th, 2023, to January 23rd, 2024 at Lille University Hospital (Lille, France). All newborns admitted to the hospital's maternity department during the study period and whose parents agreed to participate in the study were included. Parents were asked to state whether or not they agreed for their infant to receive nirsevimab. The occurrence of adverse events (AEs) 2 h after nirsevimab treatment and 7, 14 and 30 days after discharge was documented by the mother. The primary endpoint was the nirsevimab treatment acceptance rate. The secondary endpoints were the variables associated with the acceptance of nirsevimab, the reasons for accepting or refusing nirsevimab, and the treatment's real-life safety, relative to a non-treated control group of newborns. Findings Of the 1730 infants born in the hospital during the study period, 477 met all the inclusion criteria and were enrolled. The nirsevimab acceptance rate [95% confidence interval] was 91.6% [89.1%-94.2%]. In a multivariable analysis, the mother's age, lower parity and having a partner in work were significantly associated with nirsevimab acceptance. The most common reason for accepting treatment was "to protect my baby", and the most common reason for refusing treatment was the lack of long-term data on nirsevimab. The nirsevimab and control groups did not differ significantly in terms of the types and frequencies of AEs. At least one serious AE was reported for 9.4% of the infants in the nirsevimab group and for 10.3% in the control group. None of the serious AEs were considered to be related to nirsevimab treatment. Interpretation The nirsevimab acceptance rate for newborns in the maternity unit was high during the first season of implementation. The safety profile was very good, with no significant differences between the nirsevimab group and the control group. Funding None.
Collapse
Affiliation(s)
| | - Clara Testard
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
| | - Marion Lagrée
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
| | - Maxime Leroy
- CHU Lille, Département de Biostatistiques, F-59000, Lille, France
| | - Lisa Gasnier
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
| | - Alicia Enes-Dias
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
| | - Constance Leruste
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
| | - Diariatou Diallo
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
| | - Michael Génin
- CHU Lille, Département de Biostatistiques, F-59000, Lille, France
- Univ. Lille, ULR2694–METRICS: Évaluation des Technologies de Santé et des Pratiques Médicales, F-59000, Lille, France
| | - Thameur Rakza
- CHU Lille, Service de Pédiatrie de Maternité, Jeanne de Flandre Hospital, F-59000, Lille, France
| | - François Dubos
- CHU Lille, Urgences Pédiatriques & Maladies Infectieuses, Roger Salengro Hospital, F-59000, Lille, France
- Univ. Lille, ULR2694–METRICS: Évaluation des Technologies de Santé et des Pratiques Médicales, F-59000, Lille, France
| |
Collapse
|
12
|
Pérez Martín JJ, Zornoza Moreno M. Implementation of the first respiratory syncytial (RSV) immunization campaign with nirsevimab in an autonomous community in Spain. Hum Vaccin Immunother 2024; 20:2365804. [PMID: 39137331 PMCID: PMC11188787 DOI: 10.1080/21645515.2024.2365804] [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: 04/09/2024] [Accepted: 06/05/2024] [Indexed: 08/15/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the main cause of low respiratory tract infections in infants under one year of age. In the 2023/2024 season, the monoclonal antibody nirsevimab was available to protect children from RSV, and Spain has become one of the first countries worldwide to implement this strategy. It is essential to evaluate the results of this first campaign and different characteristics of the immunized population in order to plan next campaigns, especially for countries that are going to include this immunization. Our coverage was high (91.5% for those born during the season and 88.3% globally). For those born during the season, only 4.9% preferred not to immunize at the maternity hospital, which meant an average delay of 27.45 days. We observed a lower coverage in the population of immigrant origin. There was a rapid pace of immunization, since for those born before the beginning of the campaign the mean to be immunized was 15.63 days, without differences between healthy and at-risk children. This allows immunization before the RSV season (90% of the catch-up children had been immunized on November 3). The average age at which all the immunized children have received nirsevimab was lower in healthy children compared to those with risk conditions (49.65 versus 232.85 days). For those born during the campaign, the average age was also lower in healthy children (3.14 versus 14.58 days). In conclusion, we consider that the implementation of the immunization strategy with nirsevimab in the Region of Murcia, Spain, has been a success.
Collapse
|
13
|
White JT, Terstappen J, Levi M, Radivojevic A, Noble R, Anderson AB, Wise-Blackman G, Dunne MW. Replacing serum with dried blood microsampling for pharmacokinetics, viral neutralisation and immunogenicity bioanalysis supporting future paediatric development of RSM01, a candidate respiratory syncytial virus neutralising monoclonal antibody. BMC Infect Dis 2024; 24:1403. [PMID: 39696004 PMCID: PMC11653551 DOI: 10.1186/s12879-024-10196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/07/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Virus neutralising antibodies in serum are considered key correlates of protection for vaccines and monoclonal antibodies against respiratory syncytial virus (RSV). RSM01 is a novel, highly-potent, half-life-extended and fully-human monoclonal antibody candidate targeting the RSV prefusion F protein. Currently in Phase 1 development, RSM01 is primarily being developed to potentially provide an effective and affordable RSV prevention strategy in low- and middle-income countries. To evaluate the ability of dried blood collection to generate data sets and conclusions comparable to serum collection, we compared pharmacokinetics (PK) of RSM01, immunogenicity, and virus neutralisation for dried capillary blood samples with serum samples. METHODS RSM01 PK, anti-drug antibodies (ADA), and RSV-neutralising antibodies from the Phase 1 trial were analyzed and compared between matched serum and dried blood samples. Deming regression analysis was performed using baseline-corrected values to evaluate correlation between measurements in liquid serum versus dried blood. RESULTS The analysis showed good correlation (R2 > 0.95) between individual RSM01 concentrations measured in both serum and capillary blood. Analysis of RSM01 PK parameters in capillary blood yielded equivalent conclusions as from serum. A strong correlation (R2 > 0.95) was observed between RSV neutralising activity measured in both serum and capillary blood. In addition, RSV neutralising activity was correlated with RSM01 concentrations in both serum and capillary blood data sets. For ADA, individual sample results had 96% agreement (290/302) and overall participant ADA status had 93% agreement (52/56). CONCLUSIONS Both RSM01 concentrations and RSV neutralising activity showed a strong correlation between the serum and blood measurements. ADA measurements also had an agreement of > 90% for individual samples and overall participant status. Our results demonstrate that dried blood is a suitable specimen type for collection and evaluation in the RSM01 clinical development program and shows promise as a useful approach to reduce patient burden in clinical trials, particularly for infants in low- and middle-income countries. TRIAL REGISTRATION Clinicaltrials.gov NCT05118386 November 12, 2021.
Collapse
MESH Headings
- Humans
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/blood
- Respiratory Syncytial Virus Infections/prevention & control
- Dried Blood Spot Testing/methods
- Respiratory Syncytial Virus, Human/immunology
- Antibodies, Viral/blood
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/blood
- Infant
- Male
- Female
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/blood
Collapse
Affiliation(s)
- Joleen T White
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA.
| | - Jonne Terstappen
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Micha Levi
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | - Robert Noble
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | | | - Michael W Dunne
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| |
Collapse
|
14
|
Slein MD, Backes IM, Kelkar NS, Garland CR, Khanwalkar US, Sholukh AM, Johnston CM, Leib DA, Ackerman ME. Improving antibody-mediated protection against HSV infection by eliminating interactions with the viral Fc receptor gE/gI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624598. [PMID: 39605495 PMCID: PMC11601663 DOI: 10.1101/2024.11.20.624598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules, allowing the virus to escape immune clearance. In addition to their role in neuropathogenesis and cell-cell spread, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG and promote evasion of humoral immune responses. While monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of the vFcR on mAb-mediated protection by binding to IgG is unknown. Using HSV-1 with intact and ablated gE-mediated IgG Fc binding, and Fc-engineered antibodies with modified ability to interact with gE/gI, we investigated the role of the vFcR in viral pathogenesis and mAb-mediated protection from nHSV. The gD-specific human mAb HSV8 modified to lack binding to gE exhibited enhanced neutralization and in vivo protection compared to its native IgG1 form. This improved protection by the engineered mAbs was dependent on the presence of the vFcR. Human IgG3 allotypes lacking vFcR binding also exhibited enhanced antiviral activity in vivo, suggesting that vaccines that robustly induce IgG3 responses could show enhanced protection. suggesting the value of vaccination strategies that robustly induce this subclass. Lastly, analysis of longitudinal responses to acute primary genital infection in humans raised the possibility that unlike most viruses, HSV may exhibited slow induction of IgG3. In summary, this study demonstrates that mAbs lacking the ability to interact with the vFcR can exhibit improved protection from HSV-offering new prospects for antibody-based interventions.
Collapse
Affiliation(s)
- Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | | | - Christine M. Johnston
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Departments of Medicine and Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
15
|
Schwartz D, Tse-Chang A, Robinson J, Gratrix J, Smyczek P, Hawkes MT. Kinetics of RPR Decline in Pregnant Persons Treated for Syphilis in Pregnancy and Their Infants. Pathogens 2024; 13:1010. [PMID: 39599563 PMCID: PMC11597520 DOI: 10.3390/pathogens13111010] [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: 09/13/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Congenital syphilis is a re-emerging infectious threat in areas of North America. The purpose of this study was to quantitatively describe the rate of decline of nontreponemal (rapid plasma reagin, RPR) titers in pregnant persons with syphilis and their infants. In a retrospective review, we included 120 pregnant persons with 563 reactive RPR measurements (median 5, range 2 to 11 per person) and 35 infants with 81 RPR measurements (median 2, range 2 to 6 per infant). First-order decay, second-order decay, and a mathematical model representing functional FcRn-mediated antibody recycling were fitted to individual patient RPR trajectories. The RPR titers decreased with a median half-life of 39 days (IQR 28-59) and 27 days (IQR 17-41) in birthing parents and infants, respectively. The half-life varied with the initial RPR titer, suggesting that the kinetics of RPR decline was not first-order. A mathematical model accounting for saturable antibody recycling explained the longevity of RPR reactivity, predicted the observed non-linear kinetics, and fit the empiric data well. In summary, RPR titers decline with a half-life of roughly one month; however, the elimination does not follow first-order kinetics. Saturable antibody recycling may explain the prolonged and non-linear elimination of nontreponemal antibodies.
Collapse
Affiliation(s)
- Danielle Schwartz
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Alena Tse-Chang
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada; (A.T.-C.)
| | - Joan Robinson
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada; (A.T.-C.)
| | - Jennifer Gratrix
- Sexually Transmitted Infections Services, Provincial, Population, and Public Health, Alberta Health Services, Edmonton, AB T5J 3E4, Canada; (J.G.)
| | - Petra Smyczek
- Sexually Transmitted Infections Services, Provincial, Population, and Public Health, Alberta Health Services, Edmonton, AB T5J 3E4, Canada; (J.G.)
| | - Michael T. Hawkes
- Department of Pediatrics, University of British Columbia, A5-179, 950 West 28th Ave., Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
16
|
Wilkins D, Wählby Hamrén U, Chang Y, Clegg LE, Domachowske J, Englund JA, Muller WJ, Leach A, Kelly EJ, Villafana T. RSV Neutralizing Antibodies Following Nirsevimab and Palivizumab Dosing. Pediatrics 2024; 154:e2024067174. [PMID: 39350745 DOI: 10.1542/peds.2024-067174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Data describing respiratory syncytial virus (RSV) neutralizing antibody (nAb) levels for nirsevimab, a recently approved, extended half-life, anti-RSV fusion protein (F protein) monoclonal antibody, relative to the previous standard of care, palivizumab, have not been reported. METHODS MEDLEY was a randomized, palivizumab-controlled, phase 2/3 study of nirsevimab during 2 RSV seasons (season 1 and 2) in infants born preterm (≤35 weeks' gestational age; dosed season 1 only) or with congenital heart disease or chronic lung disease of prematurity (dosed seasons 1 and 2). Participants were randomly assigned to receive a single dose of nirsevimab followed by 4 monthly placebo doses, or 5 once-monthly doses of palivizumab. Anti-RSV F protein serology (ie, levels of prefusion [pre-F]/postfusion [post-F] conformation antibodies), nirsevimab and palivizumab concentrations, and RSV nAbs were measured in participant serum collected at baseline (pre-dose) and days 31, 151, and 361. RESULTS Serologic data were similar in seasons 1 and 2. Nirsevimab predominately conferred pre-F antibodies, whereas palivizumab conferred pre-F and post-F antibodies. Nirsevimab and palivizumab serum concentrations highly correlated with nAb levels in both seasons. In season 1, nAb levels in nirsevimab recipients were highest in day 31 samples and gradually declined but remained 17-fold above baseline at day 361. nAb levels in palivizumab recipients increased incrementally with monthly doses to day 151. nAb levels followed similar patterns in season 2. nAb levels were ∼10-fold higher with nirsevimab compared with palivizumab across both seasons. CONCLUSIONS Nirsevimab prophylaxis confers ∼10-fold higher and more sustained RSV nAb levels relative to palivizumab.
Collapse
Affiliation(s)
- Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Yue Chang
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D
| | | | - Joseph Domachowske
- Department of Pediatrics, State University of New York Upstate Medical University, Syracuse, New York
| | - Janet A Englund
- Department of Pediatrics, Seattle Children's Hospital Research Institute, University of Washington, Seattle, Washington
| | - William J Muller
- Ann & Robert H Lurie Children's Hospital of Chicago and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
17
|
Fuchs J, Hübner J, Schmidt A, Irrgang P, Maier C, Vieira Antão A, Oltmanns F, Thirion C, Lapuente D, Tenbusch M. Evaluation of adenoviral vector Ad19a encoding RSV-F as novel vaccine against respiratory syncytial virus. NPJ Vaccines 2024; 9:205. [PMID: 39472590 PMCID: PMC11522487 DOI: 10.1038/s41541-024-01001-z] [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: 04/10/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in infants and toddlers. Since natural infections do not induce persistent immunity, there is the need of vaccines providing long-term protection. Here, we evaluated a new adenoviral vector (rAd) vaccine based on the rare serotype rAd19a and compared the immunogenicity and efficacy to the highly immunogenic rAd5. Given as an intranasal boost in DNA primed mice, both vectors encoding the F protein provided efficient protection against a subsequent RSV infection. However, intramuscular immunization with rAd19a vectors provoked vaccine-enhanced disease after RSV infection compared to non-vaccinated animals. While mucosal IgA antibodies and tissue-resident memory T-cells in intranasally vaccinated mice rapidly control RSV replication, a strong anamnestic systemic T-cell response in absence of local immunity might be the reason for immune-mediated enhanced disease. Our study highlighted the potential benefits of developing effective mucosal against respiratory pathogens.
Collapse
Affiliation(s)
- Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Julian Hübner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Anna Schmidt
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Ana Vieira Antão
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany.
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054, Erlangen, Germany.
| |
Collapse
|
18
|
Esposito GL, Fassio F, Girardi D, Picasso E, Meloni F, Montini S, Codullo V, Pattonieri EF, Defrancesco I, Bianchessi A, Calvi M, Seminari EM, Baldanti F, Lilleri D, Novelli V, Marena C. "REAl LIfe" observational study on the effectiveness of Evusheld prophylaxis against SARS-CoV-2 omicron variants in vaccine non-responder immunocompromised patients (REALISE). Vaccine 2024; 42:126208. [PMID: 39154513 DOI: 10.1016/j.vaccine.2024.126208] [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: 03/17/2024] [Revised: 07/26/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Infection by SARS-CoV2 has become a challenge, especially for immunocompromised patients who show a weaker humoral response to COVID-19 vaccine. Tixagevimab+cilgavimab (Evusheld) is a combination of human monoclonal antibodies that can be used for pre-exposure prophylaxis to prevent infection or disease by SARS-CoV2. OBJECTIVES Our study aimed to investigate the effectiveness of Evusheld by comparing an Exposed and an Unexposed group. STUDY DESIGN Immunocompromised patients were enrolled in the Evusheld Group between March and September 2022. All patients had anti-spike IgG antibody levels <260 BAU/ml before administration of Evusheld. Blood samples for serological evaluations were collected, and anti-Spike antibodies were tested. For the Unexposed Group, a serologic test was performed at enrollment and a questionnaire was performed after 6 months. RESULTS 43 patients received Evusheld pre-exposure prophylaxis and 45 patients not receiving Evusheld were enrolled in the Unexposed group. The median age was 59.0 years in the Evusheld group, and 63.0 in the unexposed group. In the Evusheld group, during the Omicron wave in Italy, 23.3% of subjects developed symptomatic infection compared to 42.2% in the unexposed group. A majority of infections was seen in male respect to female patients. No difference in length of infection between the groups was seen. Antibody level remained higher than the basal threshold at 180 days from enrollment. CONCLUSIONS Evusheld seems to reduce the rate of symptomatic infection in immunocompromised patients. Further data are required to determine whether this prophylaxis may have a longer-lasting effect over time.
Collapse
Affiliation(s)
- Giuliana Lucia Esposito
- Fondazione IRCCS Policlinico San Matteo, Microbiology and Virology Unit, Pavia, Italy,; Specialization School in Microbiology and Virology,University of Pavia, Pavia, Italy..
| | - Federico Fassio
- Department of Public Health, Experimental and Forensic Medicine, Section of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia,Italy; Fondazione IRCCS Policlinico San Matteo, Medical direction, Pavia, Italy
| | - Daniela Girardi
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Italy.; Fondazione IRCCS Policlinico San Matteo, Medical direction, Pavia, Italy
| | - Erica Picasso
- Fondazione IRCCS Policlinico San Matteo, Medical direction, Pavia, Italy
| | - Federica Meloni
- Fondazione IRCCS Policlinico San Matteo, UOS transplant center, Pavia, Italy
| | - Simone Montini
- Fondazione IRCCS Policlinico San Matteo, UOS transplant center, Pavia, Italy
| | - Veronica Codullo
- Fondazione IRCCS Policlinico San Matteo, Division of Rheumatology, Pavia, Italy
| | | | - Irene Defrancesco
- Fondazione IRCCS Policlinico San Matteo, Division of Haematology, Pavia, Italy; Department of surgical, clinical, diagnostic and pediatric sciences, University of Pavia, Pavia, Italy
| | - Antonio Bianchessi
- Fondazione IRCCS Policlinico San Matteo, Division of Haematology, Pavia, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Monica Calvi
- Fondazione IRCCS Policlinico San Matteo, Pharmacy Unit, Pavia, Italy
| | - Elena Maria Seminari
- Fondazione IRCCS Policlinico San Matteo, Infectious diseases division, Pavia, Italy
| | - Fausto Baldanti
- Fondazione IRCCS Policlinico San Matteo, Microbiology and Virology Unit, Pavia, Italy,; Department of surgical, clinical, diagnostic and pediatric sciences, University of Pavia, Pavia, Italy
| | - Daniele Lilleri
- Fondazione IRCCS Policlinico San Matteo, Microbiology and Virology Unit, Pavia, Italy
| | - Viola Novelli
- Fondazione IRCCS Policlinico San Matteo, Medical direction, Pavia, Italy
| | - Carlo Marena
- Fondazione IRCCS Policlinico San Matteo, Medical direction, Pavia, Italy
| |
Collapse
|
19
|
Loe MWC, Soenong H, Lee E, Li-Kim-Moy J, Williams PC, Yeo KT. Nirsevimab: Alleviating the burden of RSV morbidity in young children. J Paediatr Child Health 2024; 60:489-498. [PMID: 39150043 DOI: 10.1111/jpc.16643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections (LRTIs) and hospital admissions in early childhood. Recent advancements in novel preventive therapies, including extended half-life monoclonal antibodies and antenatal vaccination, have afforded new opportunities to significantly reduce the burden of this infection. Nirsevimab is a novel monoclonal antibody that provides sustained protection against RSV for at least 5 months among newborns and young children. It has received regulatory approval in numerous countries and is being implemented across various settings. Two pivotal Phase 3 trials (MELODY, HARMONIE) demonstrated significant reductions in RSV-associated LRTI hospitalisations following nirsevimab administration, with treatment efficacy of 62.1% and 83.2%. Emerging real-world data from early adopters of nirsevimab corroborates these findings. Studies from Spain, Luxembourg, France and the USA report effectiveness rates between 82% and 90% in preventing RSV-associated hospitalisations among infants entering their first RSV season. Current implementation strategies for nirsevimab have primarily focused on seasonal administration for all infants, aligned to local RSV seasons, and often include catch-up doses for those born before the season begins. Available cost-effectiveness analyses indicate that while nirsevimab offers significant potential public health benefits, its adoption must carefully consider economic factors such as treatment costs, implementation strategies tailored to local viral epidemiology, and logistics for vaccine delivery. Overall, nirsevimab presents a promising opportunity to alleviate the burden of severe RSV infections in young children. However, ongoing surveillance and refinements in implementation strategies are crucial to optimise its impact and ensure sustainability across diverse health-care settings.
Collapse
Affiliation(s)
- Marcus Wing Choy Loe
- Duke-NUS Medicine School, Singapore
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
| | - Helen Soenong
- School of Women's and Children's Health, University of New South Wales, Randwick, New South Wales, Australia
| | - Evelyn Lee
- Centre for Economic Impacts of Genomic Medicine, Macquarie University, Sydney, New South Wales, Australia
- Centre for Social Research in Health, University of New South Wales, Sydney, Australia
| | - Jean Li-Kim-Moy
- National Centre for Immunisation Research and Surveillance, Westmead, New South Wales, Australia
- School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Phoebe Cm Williams
- School of Women's and Children's Health, University of New South Wales, Randwick, New South Wales, Australia
- National Centre for Immunisation Research and Surveillance, Westmead, New South Wales, Australia
- School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Department of Infectious Diseases, Sydney Children's Hospital Network, Sydney, New South Wales, Australia
| | - Kee Thai Yeo
- Duke-NUS Medicine School, Singapore
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
- School of Women's and Children's Health, University of New South Wales, Randwick, New South Wales, Australia
| |
Collapse
|
20
|
Domachowske J, Hamrén UW, Banu I, Baronio R, Basavaraju B, Koen A, Leach A, Mankad VS, Pannaraj PS, Soler-Palacin P, Takas T, Mori M, Villafana T. Safety and Pharmacokinetics of Nirsevimab in Immunocompromised Children. Pediatrics 2024; 154:e2024066508. [PMID: 39257380 DOI: 10.1542/peds.2024-066508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Immunocompromised children may have increased risk for severe respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI), potentially leading to prolonged hospitalization, intensive care, and death. The open-label phase II MUSIC trial evaluated the safety and pharmacokinetics of nirsevimab, an extended half-life monoclonal antibody against RSV, in immunocompromised children aged ≤24 months. METHODS Participants received a single intramuscular injection of nirsevimab (first RSV season: 50 mg if <5 kg/100 mg if ≥5 kg; second season: 200 mg). Safety, antidrug antibodies, and pharmacokinetics were evaluated to day 361. RESULTS Participants (n = 100) had ≥1 immunocompromising conditions: primary immunodeficiency (n = 33), previous transplantation (n = 16), HIV infection (n = 8) or treatment with high-dose systemic corticosteroids (n = 29), immunosuppressive chemotherapy (n = 20), or other immunosuppressive therapies (n = 15). Six children experienced eight treatment-related adverse events (none categorized as serious). Three deaths occurred, all were unrelated to treatment. Eleven children, developed antidrug antibodies, with minimal effects on pharmacokinetics and no apparent impact on safety. Nirsevimab serum concentrations at day 151 were similar to those effective in preventing medically attended RSV LRTI in healthy infants. Fourteen children had increased nirsevimab clearance. No protocol-defined medically attended RSV LRTIs occured through day 151. CONCLUSIONS Among immunocompromised children aged ≤24 months, nirsevimab was well tolerated with no safety concerns and serum concentrations were supportive of efficacy. A subset of children with increased nirsevimab clearance, had conditions potentially associated with protein loss; however, the impact on efficacy is unknown.
Collapse
Affiliation(s)
- Joseph Domachowske
- State University of New York Upstate Medical University Syracuse, New York
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Irfana Banu
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Roberta Baronio
- Biometrics, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Bhanu Basavaraju
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Anthonet Koen
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (Wits-VIDA), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amanda Leach
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Vaishali S Mankad
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Durham, North Carolina
| | - Pia S Pannaraj
- University of California San Diego, San Diego, California
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d'Hebron, and Vall d'Hebron Research Institute, Barcelona, Catalonia, Spain
| | - Therese Takas
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Masaaki Mori
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tonya Villafana
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
21
|
Banoun H. Analysis of Beyfortus ® (Nirsevimab) Immunization Campaign: Effectiveness, Biases, and ADE Risks in RSV Prevention. Curr Issues Mol Biol 2024; 46:10369-10395. [PMID: 39329969 PMCID: PMC11431526 DOI: 10.3390/cimb46090617] [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: 07/27/2024] [Revised: 08/27/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Respiratory infections with respiratory syncytial virus (RSV) account for an important part of hospital admissions for acute respiratory infections. Nirsevimab has been developed to reduce the hospital burden of RSV infections. Compared with the product previously used, it has a stronger binding capacity to RSV F protein and a high affinity for FcRn (neonatal receptor for the Fc fragment of IgG), which extends its lifespan. Nirsevimab has been shown to be highly effective in reducing hospitalization rates of RSV infections but a large or unknown number of treated subjects have been excluded in clinical and post-marketing studies. However, analysis of these studies cannot exclude that, in rare cases, nirsevimab facilitates and worsens RSV infection (or other respiratory infections). This could be attributable to antibody-dependent enhancement (ADE) which has been observed with RSV F protein antibodies in inactivated vaccine trials. This risk has been incompletely assessed in pre-clinical and clinical trials (incomplete exploration of nirsevimab effector functions and pharmacokinetics). ADE by disruption of the immune system (not studied and due to FcRn binding) could explain why there is no reduction in all-cause hospital admissions in treated age groups. Given the high price of nirsevimab, the cost-effectiveness of mass immunization campaigns may therefore be debated from an economic as well as a scientific point of view.
Collapse
|
22
|
Peng R, Chen C, Chen Q, Zhang Y, Huang R, Zhang Y, Li J. Global progress in clinical research on human respiratory syncytial virus vaccines. Front Microbiol 2024; 15:1457703. [PMID: 39286350 PMCID: PMC11402711 DOI: 10.3389/fmicb.2024.1457703] [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: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Human respiratory syncytial virus (hRSV) not only affects newborns but also older adults, contributing to a substantial worldwide burden of disease. However, only three approved hRSV vaccines remain commercially available to date. The development of a safe, practical and broad-spectrum vaccine suitable for all age groups remains extremely challenging. Using five different approaches-live-attenuated, recombinant-vector, subunit, particle-based, and mRNA-nearly 30 hRSV vaccine candidates are currently conducting clinical trials worldwide; moreover, > 30 vaccines are under preclinical evaluation. This review presents a comprehensive overview of these hRSV vaccines along with prospects for the development of infectious disease vaccines in the post-COVID-19 pandemic era.
Collapse
Affiliation(s)
- Ruofan Peng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenghao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qian Chen
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Yuwen Zhang
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Renjin Huang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianhua Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
23
|
Sevendal ATK, Hurley S, Bartlett AW, Rawlinson W, Walker GJ. Systematic Review of the Efficacy and Safety of RSV-Specific Monoclonal Antibodies and Antivirals in Development. Rev Med Virol 2024; 34:e2576. [PMID: 39209729 DOI: 10.1002/rmv.2576] [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: 03/26/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of acute respiratory infection amongst all ages, causing a significant global health burden. Preventative and therapeutic options for RSV infection have long been under development, and recently, several widely-publicised vaccines targeting older adult and maternal populations have become available. Promising monoclonal antibody (mAb) and antiviral (AV) therapies are also progressing in clinical trials, with the prophylactic mAb nirsevimab recently approved for clinical use in infant populations. A systematic review on current progress in this area is lacking. We performed a systematic literature search (PubMed, Embase, Web of Science, ClinicalTrials.gov, EudraCT, ANZCTR-searched Nov 29th, 2023) to identify studies on all RSV-specific mAbs and AV therapies that has undergone human clinical trials since year 2000. Data extraction focused on outcomes related to the therapeutic efficacy and safety of the intervention on trial, and all studies were graded against the OCEBM Levels of Evidence Table. Results from 59 studies were extracted, covering efficacy and safety data on six mAbs (motavizumab, motavizumab-YTE, nirsevimab, ALX-0171, suptavumab, clesrovimab) and 12 AV therapies (ALN-RSV01, RSV604, presatovir, MDT-637, lumicitabine, IFN-α1b, rilematovir, enzaplatovir, AK0529, sisunatovir, PC786, EDP-938). Of the mAbs reviewed, nirsevimab and clesrovimab hold considerable promise. The timeline for RSV-specific AV availability is less advanced, although EDP-938 and AK0529 have reported promising phase 2 efficacy and safety data. Moving forward, passive immunisation and treatment options for RSV infection will play a significant role in reducing the health burden of RSV, complementing recent advancements in vaccine development. TRIAL REGISTRATION: PROSPERO registration: CRD42022376633.
Collapse
Affiliation(s)
- Andrea T K Sevendal
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
| | - Siobhan Hurley
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
| | - Adam W Bartlett
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital Network, Sydney, Australia
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - William Rawlinson
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Gregory J Walker
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
24
|
Focosi D. Monoclonal Antibody Therapies Against SARS-CoV-2: Promises and Realities. Curr Top Microbiol Immunol 2024. [PMID: 39126484 DOI: 10.1007/82_2024_268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Monoclonal antibodies targeting the Spike protein of SARS-CoV-2 have been widely deployed in the ongoing COVID-19 pandemic. I review here the impact of those therapeutics in the early pandemic, ranging from structural classification to outcomes in clinical trials to in vitro and in vivo evidence of basal and treatment-emergent immune escape. Unfortunately, the Omicron variant of concern has completely reset all achievements so far in mAb therapy for COVID-19. Despite the intrinsic limitations of this strategy, future developments such as respiratory delivery of further engineered mAb cocktails could lead to improved outcomes.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy.
| |
Collapse
|
25
|
Santarpia G, Carnes E. Therapeutic Applications of Aptamers. Int J Mol Sci 2024; 25:6742. [PMID: 38928448 PMCID: PMC11204156 DOI: 10.3390/ijms25126742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Affinity reagents, or target-binding molecules, are quite versatile and are major workhorses in molecular biology and medicine. Antibodies are the most famous and frequently used type and they have been used for a wide range of applications, including laboratory techniques, diagnostics, and therapeutics. However, antibodies are not the only available affinity reagents and they do have significant drawbacks, including laborious and costly production. Aptamers are one potential alternative that have a variety of unique advantages. They are single stranded DNA or RNA molecules that can be selected for binding to many targets including proteins, carbohydrates, and small molecules-for which antibodies typically have low affinity. There are also a variety of cost-effective methods for producing and modifying nucleic acids in vitro without cells, whereas antibodies typically require cells or even whole animals. While there are also significant drawbacks to using aptamers in therapeutic applications, including low in vivo stability, aptamers have had success in clinical trials for treating a variety of diseases and two aptamer-based drugs have gained FDA approval. Aptamer development is still ongoing, which could lead to additional applications of aptamer therapeutics, including antitoxins, and combinatorial approaches with nanoparticles and other nucleic acid therapeutics that could improve efficacy.
Collapse
Affiliation(s)
- George Santarpia
- College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Eric Carnes
- College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
26
|
Eto T, Okubo Y, Momose A, Tamura H, Zheng R, Callendret B, Bastian A, Comeaux C. A Randomized, Double-Blind, Placebo-Controlled, Phase 1 Study to Evaluate the Safety, Reactogenicity, and Immunogenicity of Single Vaccination of Ad26.RSV.preF-Based Regimen in Japanese Adults Aged 60 Years and Older. Influenza Other Respir Viruses 2024; 18:e13336. [PMID: 38880785 PMCID: PMC11180550 DOI: 10.1111/irv.13336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is increasingly recognized as a significant cause of lower respiratory tract disease (LRTD) in older adults. The Ad26.RSV.preF/RSV preF protein vaccine demonstrated protective efficacy against RSV related LRTD in a Phase 2b study in the United States. Hence, Ad26.RSV.preF/RSV preF protein vaccine candidate was evaluated in the Japanese older adult population. METHODS This Phase 1 study evaluated safety, reactogenicity, and immunogenicity of Ad26.RSV.preF/RSV preF protein vaccine at dose level of 1 × 1011 vp/150 μg in Japanese healthy adult aged ≥60 years. The study included a screening Phase, vaccination, 28-day follow up Phase, a 182-day follow-up period, and final visit on Day 183. A total of 36 participants were randomized in a 2:1 ratio to receive Ad26.RSV.preF/RSV preF protein vaccine (n = 24) or placebo (n = 12). After study intervention administration, the safety and immunogenicity analysis were performed as per planned schedule. Immune responses including virus-neutralizing and preF-specific binding antibodies were measured on Days 1, 15, 29, and 183. RESULTS There were no deaths, SAEs, or AEs leading to discontinuation reported during the study. The Ad26.RSV.preF/RSV preF protein vaccine had acceptable safety and tolerability profile with no safety concern in Japanese older adults. The Ad26.RSV.preF/RSV preF protein vaccine induced RSV-specific humoral immunity, with increase in antibody titers on Days 15 and 29 compared with baseline which was well maintained until Day 183. CONCLUSIONS A single dose of Ad26.RSV.preF/RSV preF protein vaccine had an acceptable safety and tolerability profile and induced RSV-specific humoral immunity in Japanese healthy adults. TRIAL REGISTRATION NCT number: NCT04354480; Clinical Registry number: CR108768.
Collapse
|
27
|
Clegg L, Freshwater E, Leach A, Villafana T, Hamrén UW. Population Pharmacokinetics of Nirsevimab in Preterm and Term Infants. J Clin Pharmacol 2024; 64:555-567. [PMID: 38294353 DOI: 10.1002/jcph.2401] [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: 10/19/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024]
Abstract
Nirsevimab, a monoclonal antibody with an extended half-life, is approved for the prevention of respiratory syncytial virus (RSV) disease in all infants in Canada, the EU, Great Britain, and the USA. A population pharmacokinetics (PK) model was built to describe the PK of nirsevimab in preterm and term infants, and to evaluate the influence of covariates, including body weight and age, in infants. Nirsevimab PK was characterized by a 2-compartment model with first-order clearance (CL) and first-order absorption following intramuscular (IM) administration. The typical CL in a 5 kg infant was 3.4 mL/day. Body weight and postmenstrual age were the primary covariates on CL, with minor effects for race, second RSV season, and antidrug antibody status (deemed not clinically relevant). Congenital heart disease (CHD) and chronic lung disease (CLD) did not significantly impact nirsevimab PK. The final population PK model, based on 8987 PK observations from 2683 participants across 5 clinical trials, successfully predicted PK in an additional cohort of 967 healthy infants. Weight-banded dosing (50 mg in infants <5 kg; 100 mg in infants ≥5 kg) was predicted to be appropriate for infants ≥1 kg in their first RSV season. Together, these data support weight-banded dosing of nirsevimab in all infants in their first RSV season, including in healthy infants, infants with CHD or CLD, and in infants born prematurely.
Collapse
Affiliation(s)
- Lindsay Clegg
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Amanda Leach
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
28
|
Kim H, Taslakjian B, Kim S, Tirrell MV, Guler MO. Therapeutic Peptides, Proteins and their Nanostructures for Drug Delivery and Precision Medicine. Chembiochem 2024; 25:e202300831. [PMID: 38408302 DOI: 10.1002/cbic.202300831] [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: 12/08/2023] [Revised: 02/05/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Peptide and protein nanostructures with tunable structural features, multifunctionality, biocompatibility and biomolecular recognition capacity enable development of efficient targeted drug delivery tools for precision medicine applications. In this review article, we present various techniques employed for the synthesis and self-assembly of peptides and proteins into nanostructures. We discuss design strategies utilized to enhance their stability, drug-loading capacity, and controlled release properties, in addition to the mechanisms by which peptide nanostructures interact with target cells, including receptor-mediated endocytosis and cell-penetrating capabilities. We also explore the potential of peptide and protein nanostructures for precision medicine, focusing on applications in personalized therapies and disease-specific targeting for diagnostics and therapeutics in diseases such as cancer.
Collapse
Affiliation(s)
- HaRam Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Boghos Taslakjian
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Sarah Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Matthew V Tirrell
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| |
Collapse
|
29
|
Levien TL, Baker DE. Formulary Drug Reviews: Nirsevimab. Hosp Pharm 2024; 59:138-145. [PMID: 38450347 PMCID: PMC10913880 DOI: 10.1177/00185787231212620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Each month, subscribers to The Formulary Monograph Service receive 5 to 6 well-documented monographs on drugs that are newly released or are in late phase 3 trials. The monographs are targeted to Pharmacy & Therapeutics Committees. Subscribers also receive monthly 1-page summary monographs on agents that are useful for agendas and pharmacy/nursing in-services. A comprehensive target drug utilization evaluation/medication use evaluation (DUE/MUE) is also provided each month. With a subscription, the monographs are available online to subscribers. Monographs can be customized to meet the needs of a facility. Through the cooperation of The Formulary, Hospital Pharmacy publishes selected reviews in this column. For more information about The Formulary Monograph Service, contact Wolters Kluwer customer service at 866-397-3433.
Collapse
|
30
|
McPherson C, Lockowitz CR, Newland JG. Balanced on the Biggest Wave: Nirsevimab for Newborns. Neonatal Netw 2024; 43:105-115. [PMID: 38599778 DOI: 10.1891/nn-2023-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of hospitalization in infancy in the United States. Nearly all infants are infected by 2 years of age, with bronchiolitis requiring hospitalization often occurring in previously healthy children and long-term consequences of severe disease including delayed speech development and asthma. Incomplete passage of maternal immunity and a high degree of genetic variability within the virus contribute to morbidity and have also prevented successful neonatal vaccine development. Monoclonal antibodies reduce the risk of hospitalization from severe RSV disease, with palivizumab protecting high-risk newborns with comorbidities including chronic lung disease and congenital heart disease. Unfortunately, palivizumab is costly and requires monthly administration of up to five doses during the RSV season for optimal protection.Rapid advances in the past two decades have facilitated the identification of antibodies with broad neutralizing activity and allowed manipulation of their genetic code to extend half-life. These advances have culminated with nirsevimab, a monoclonal antibody targeting the Ø antigenic site on the RSV prefusion protein and protecting infants from severe disease for an entire 5-month season with a single dose. Four landmark randomized controlled trials, the first published in July 2020, have documented the efficacy and safety of nirsevimab in healthy late-preterm and term infants, healthy preterm infants, and high-risk preterm infants and those with congenital heart disease. Nirsevimab reduces the risk of RSV disease requiring medical attention (number needed to treat [NNT] 14-24) and hospitalization (NNT 33-63) with rare mild rash and injection site reactions. Consequently, the Centers for Disease Control and Prevention has recently recommended nirsevimab for all infants younger than 8 months of age entering or born during the RSV season and high-risk infants 8-19 months of age entering their second season. Implementing this novel therapy in this large population will require close multidisciplinary collaboration. Equitable distribution through minimizing barriers and maximizing uptake must be prioritized.
Collapse
|
31
|
Yu T, Padula WV, Yieh L, Gong CL. Cost-effectiveness of nirsevimab and palivizumab for respiratory syncytial virus prophylaxis in preterm infants 29-34 6/7 weeks' gestation in the United States. Pediatr Neonatol 2024; 65:152-158. [PMID: 37758594 DOI: 10.1016/j.pedneo.2023.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/31/2023] [Accepted: 04/26/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) hospitalizations have increased since the 2014 guideline update recommended against the use of palivizumab for preterm infants born ≥29 0/7 weeks' gestational age (GA) without additional risk factors. A novel drug candidate, nirsevimab, has been developed for this population. We analyzed the cost-effectiveness of palivizumab/nirsevimab vs. no prophylaxis in this population. METHODS A hybrid-Markov model predicted the RSV clinical course in the first year of life and sequelae in the subsequent four years for preterm infants from the healthcare and societal perspectives. Model parameters were derived from the literature. We calculated costs and quality-adjusted life-years (QALYs) to produce an incremental cost-effectiveness ratio (ICER) evaluated at a willingness-to-pay threshold of $150,000/QALY. Sensitivity analyses assessed model robustness. A threshold analysis examined nirsevimab pricing uncertainty. RESULTS Compared to no prophylaxis, palivizumab costs $9572 and $9584 more from the healthcare and societal perspectives, respectively, with 0.0019 QALYs gained per patient over five years, resulting in ICERs >$5 million per QALY from each perspective. Results were robust to parameter uncertainties; probabilistic sensitivity analysis revealed that no prophylaxis had a 100% probability of being cost-effective. The threshold analysis suggested that nirsevimab is not cost-effective when compared to no prophylaxis if the price exceeds $1962 from a societal perspective. CONCLUSION Palivizumab is dominated by no prophylaxis for preterm infants 29 0/7-34 6/7 weeks' GA with no additional risk factors. Relevant stakeholders should consider alternatives to palivizumab for this population that are both effective and economical.
Collapse
Affiliation(s)
- Tianzhou Yu
- Department of Pharmaceutical and Health Economics, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA.
| | - William V Padula
- Department of Pharmaceutical and Health Economics, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA; Leonard D. Schaeffer Center for Health Policy and Economics, Mann School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Leah Yieh
- Leonard D. Schaeffer Center for Health Policy and Economics, Mann School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Fetal & Neonatal Institute, Division of Neonatology, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Cynthia L Gong
- Leonard D. Schaeffer Center for Health Policy and Economics, Mann School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Fetal & Neonatal Institute, Division of Neonatology, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
32
|
Loaiza RA, Ramírez RA, Sepúlveda-Alfaro J, Ramírez MA, Andrade CA, Soto JA, González PA, Bueno SM, Kalergis AM. A molecular perspective for the development of antibodies against the human respiratory syncytial virus. Antiviral Res 2024; 222:105783. [PMID: 38145755 DOI: 10.1016/j.antiviral.2023.105783] [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: 10/07/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/27/2023]
Abstract
The human respiratory syncytial virus (hRSV) is the leading etiologic agent causing respiratory infections in infants, children, older adults, and patients with comorbidities. Sixty-seven years have passed since the discovery of hRSV, and only a few successful mitigation or treatment tools have been developed against this virus. One of these is immunotherapy with monoclonal antibodies against structural proteins of the virus, such as Palivizumab, the first prophylactic approach approved by the Food and Drug Administration (FDA) of the USA. In this article, we discuss different strategies for the prevention and treatment of hRSV infection, focusing on the molecular mechanisms against each target that underly the rational design of antibodies against hRSV. At the same time, we describe the latest results regarding currently approved therapies against hRSV and the challenges associated with developing new candidates.
Collapse
Affiliation(s)
- Ricardo A Loaiza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Robinson A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Mario A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
33
|
Zou G, Cao S, Gao Z, Yie J, Wu JZ. Current state and challenges in respiratory syncytial virus drug discovery and development. Antiviral Res 2024; 221:105791. [PMID: 38160942 DOI: 10.1016/j.antiviral.2023.105791] [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: 11/21/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Human respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections (LRTI) in young children and elderly people worldwide. Recent significant progress in our understanding of the structure and function of RSV proteins has led to the discovery of several clinical candidates targeting RSV fusion and replication. These include both the development of novel small molecule interventions and the isolation of potent monoclonal antibodies. In this review, we summarize the state-of-the-art of RSV drug discovery, with a focus on the characteristics of the candidates that reached the clinical stage of development. We also discuss the lessons learned from failed and discontinued clinical developments and highlight the challenges that remain for development of RSV therapies.
Collapse
Affiliation(s)
- Gang Zou
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China.
| | - Sushan Cao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Zhao Gao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Junming Yie
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Jim Zhen Wu
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| |
Collapse
|
34
|
Drysdale SB, Cathie K, Flamein F, Knuf M, Collins AM, Hill HC, Kaiser F, Cohen R, Pinquier D, Felter CT, Vassilouthis NC, Jin J, Bangert M, Mari K, Nteene R, Wague S, Roberts M, Tissières P, Royal S, Faust SN. Nirsevimab for Prevention of Hospitalizations Due to RSV in Infants. N Engl J Med 2023; 389:2425-2435. [PMID: 38157500 DOI: 10.1056/nejmoa2309189] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
BACKGROUND The safety of the monoclonal antibody nirsevimab and the effect of nirsevimab on hospitalizations for respiratory syncytial virus (RSV)-associated lower respiratory tract infection when administered in healthy infants are unclear. METHODS In a pragmatic trial, we randomly assigned, in a 1:1 ratio, infants who were 12 months of age or younger, had been born at a gestational age of at least 29 weeks, and were entering their first RSV season in France, Germany, or the United Kingdom to receive either a single intramuscular injection of nirsevimab or standard care (no intervention) before or during the RSV season. The primary end point was hospitalization for RSV-associated lower respiratory tract infection, defined as hospital admission and an RSV-positive test result. A key secondary end point was very severe RSV-associated lower respiratory tract infection, defined as hospitalization for RSV-associated lower respiratory tract infection with an oxygen saturation of less than 90% and the need for supplemental oxygen. RESULTS A total of 8058 infants were randomly assigned to receive nirsevimab (4037 infants) or standard care (4021 infants). Eleven infants (0.3%) in the nirsevimab group and 60 (1.5%) in the standard-care group were hospitalized for RSV-associated lower respiratory tract infection, which corresponded to a nirsevimab efficacy of 83.2% (95% confidence interval [CI], 67.8 to 92.0; P<0.001). Very severe RSV-associated lower respiratory tract infection occurred in 5 infants (0.1%) in the nirsevimab group and in 19 (0.5%) in the standard-care group, which represented a nirsevimab efficacy of 75.7% (95% CI, 32.8 to 92.9; P = 0.004). The efficacy of nirsevimab against hospitalization for RSV-associated lower respiratory tract infection was 89.6% (adjusted 95% CI, 58.8 to 98.7; multiplicity-adjusted P<0.001) in France, 74.2% (adjusted 95% CI, 27.9 to 92.5; multiplicity-adjusted P = 0.006) in Germany, and 83.4% (adjusted 95% CI, 34.3 to 97.6; multiplicity-adjusted P = 0.003) in the United Kingdom. Treatment-related adverse events occurred in 86 infants (2.1%) in the nirsevimab group. CONCLUSIONS Nirsevimab protected infants against hospitalization for RSV-associated lower respiratory tract infection and against very severe RSV-associated lower respiratory tract infection in conditions that approximated real-world settings. (Funded by Sanofi and AstraZeneca; HARMONIE ClinicalTrials.gov number, NCT05437510).
Collapse
Affiliation(s)
- Simon B Drysdale
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Katrina Cathie
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Florence Flamein
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Markus Knuf
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Andrea M Collins
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Helen C Hill
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Friedrich Kaiser
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Robert Cohen
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Didier Pinquier
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Christian T Felter
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Natalya C Vassilouthis
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Jing Jin
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Mathieu Bangert
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Karine Mari
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Rapi Nteene
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Sophie Wague
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Michelle Roberts
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Pierre Tissières
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Simon Royal
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| | - Saul N Faust
- From the Centre for Neonatal and Paediatric Infections, St. George's, University of London, and the Department of Paediatrics, St. George's University Hospitals National Health Service (NHS) Foundation Trust, London (S.B.D.), the National Institute for Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and the Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton (K.C., S.N.F.), the Liverpool Vaccine Group, Liverpool School of Tropical Medicine (A.M.C., H.C.H.), and Liverpool University Hospitals Foundation, NHS Trust (A.M.C.), Liverpool, Sanofi, Reading (C.T.F., N.C.V.), and the University of Nottingham Health Service, University of Nottingham, Nottingham (S.R.) - all in the United Kingdom; Université de Lille, INSERM, Centre Hospitalier Universitaire (CHU) de Lille, CIC-1403 INSERM-CHU, Lille (F.F.), the French Clinical Research Infrastructure Network-PEDSTART, Tours (F.F.), Centre Hospitalier Intercommunal de Créteil, and Association Clinique et Thérapeutique Infantile du Val-de-Marne, Créteil (R.C.), CHU Rouen, Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Charles Nicolle University Hospital, Rouen (D.P.), Sanofi Vaccines, Lyon (M.B., R.N., S.W.), Sanofi Vaccines, Marcy L'Etoile (K.M.), Pediatric Intensive Care, Neonatal Medicine and Pediatric Emergency Department, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre (P.T.), and the Institute of Integrative Biology of the Cell, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Paris-Saclay University, Gif sur Yvette (P.T.) - all in France; Children's Hospital, Worms (M.K.), Pediatric Infectious Diseases, University Medicine, Mainz (M.K.), and Gemeinschaftspraxis für Kinder und Jugendmedizin, Tangstedter Landstrasse 77, Hamburg (F.K.) - all in Germany; Sanofi, Huipu Mansion, Beijing (J.J.); and Sanofi Vaccines, Bridgewater, NJ (M.R.)
| |
Collapse
|
35
|
Yang X. Passive antibody therapy in emerging infectious diseases. Front Med 2023; 17:1117-1134. [PMID: 38040914 DOI: 10.1007/s11684-023-1021-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/20/2023] [Indexed: 12/03/2023]
Abstract
The epidemic of corona virus disease 2019 (COVID-19) caused by severe acute respiratory syndrome Coronavirus 2 and its variants of concern (VOCs) has been ongoing for over 3 years. Antibody therapies encompassing convalescent plasma, hyperimmunoglobulin, and neutralizing monoclonal antibodies (mAbs) applied in passive immunotherapy have yielded positive outcomes and played a crucial role in the early COVID-19 treatment. In this review, the development path, action mechanism, clinical research results, challenges, and safety profile associated with the use of COVID-19 convalescent plasma, hyperimmunoglobulin, and mAbs were summarized. In addition, the prospects of applying antibody therapy against VOCs was assessed, offering insights into the coping strategies for facing new infectious disease outbreaks.
Collapse
Affiliation(s)
- Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, 430207, China.
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, 430207, China.
- China National Biotec Group Company Limited, Beijing, 100029, China.
| |
Collapse
|
36
|
Abelson D, Barajas J, Stuart L, Kim D, Marimuthu A, Hu C, Yamamoto B, Ailor E, Whaley KJ, Vu H, Agans KN, Borisevich V, Deer DJ, Dobias NS, Woolsey C, Prasad AN, Peel JE, Lawrence WS, Cross RW, Geisbert TW, Fenton KA, Zeitlin L. Long-term Prophylaxis Against Aerosolized Marburg Virus in Nonhuman Primates With an Afucosylated Monoclonal Antibody. J Infect Dis 2023; 228:S701-S711. [PMID: 37474248 PMCID: PMC11009508 DOI: 10.1093/infdis/jiad278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Marburg virus (MARV) causes a hemorrhagic fever disease in human and nonhuman primates with high levels of morbidity and mortality. Concerns about weaponization of aerosolized MARV have spurred the development of nonhuman primate (NHP) models of aerosol exposure. To address the potential threat of aerosol exposure, a monoclonal antibody that binds MARV glycoprotein was tested, MR186YTE, for its efficacy as a prophylactic. MR186YTE was administered intramuscularly to NHPs at 15 or 5 mg/kg 1 month prior to MARV aerosol challenge. Seventy-five percent (3/4) of the 15 mg/kg dose group and 50% (2/4) of the 5 mg/kg dose group survived. Serum analyses showed that the NHP dosed with 15 mg/kg that succumbed to infection developed an antidrug antibody response and therefore had no detectable MR186YTE at the time of challenge. These results suggest that intramuscular dosing of mAbs may be a clinically useful prophylaxis for MARV aerosol exposure.
Collapse
Affiliation(s)
- Dafna Abelson
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Lauren Stuart
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | - Do Kim
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Chris Hu
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Eric Ailor
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Hong Vu
- Integrated Biotherapeutics, Rockville, Maryland, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jennifer E Peel
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - William S Lawrence
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| |
Collapse
|
37
|
Brady T, Cayatte C, Roe TL, Speer SD, Ji H, Machiesky L, Zhang T, Wilkins D, Tuffy KM, Kelly EJ. Fc-mediated functions of nirsevimab complement direct respiratory syncytial virus neutralization but are not required for optimal prophylactic protection. Front Immunol 2023; 14:1283120. [PMID: 37901217 PMCID: PMC10600457 DOI: 10.3389/fimmu.2023.1283120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Nirsevimab is an extended half-life (M252Y/S254T/T256E [YTE]-modified) monoclonal antibody to the pre-fusion conformation of the respiratory syncytial virus (RSV) Fusion protein, with established efficacy in preventing RSV-associated lower respiratory tract infection in infants for the duration of a typical RSV season. Previous studies suggest that nirsevimab confers protection via direct virus neutralization. Here we use preclinical models to explore whether fragment crystallizable (Fc)-mediated effector functions contribute to nirsevimab-mediated protection. Methods Nirsevimab, MEDI8897* (i.e., nirsevimab without the YTE modification), and MEDI8897*-TM (i.e., MEDI8897* without Fc effector functions) binding to Fc γ receptors (FcγRs) was evaluated using surface plasmon resonance. Antibody-dependent neutrophil phagocytosis (ADNP), antibody-dependent cellular phagocytosis (ADCP), antibody-dependent complement deposition (ADCD), and antibody-dependent cellular cytotoxicity (ADCC) were assessed through in vitro and ex vivo serological analyses. A cotton rat challenge study was performed with MEDI8897* and MEDI8897*-TM to explore whether Fc effector functions contribute to protection from RSV. Results Nirsevimab and MEDI8897* exhibited binding to a range of FcγRs, with expected reductions in FcγR binding affinities observed for MEDI8897*-TM. Nirsevimab exhibited in vitro ADNP, ADCP, ADCD, and ADCC activity above background levels, and similar ADNP, ADCP, and ADCD activity to palivizumab. Nirsevimab administration increased ex vivo ADNP, ADCP, and ADCD activity in participant serum from the MELODY study (NCT03979313). However, ADCC levels remained similar between nirsevimab and placebo. MEDI8897* and MEDI8897*-TM exhibited similar dose-dependent reduction in lung and nasal turbinate RSV titers in the cotton rat model. Conclusion Nirsevimab possesses Fc effector activity comparable with the current standard of care, palivizumab. However, despite possessing the capacity for Fc effector activity, data from RSV challenge experiments illustrate that nirsevimab-mediated protection is primarily dependent on direct virus neutralization.
Collapse
Affiliation(s)
- Tyler Brady
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Corinne Cayatte
- Early Oncology ICA, Oncology R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Tiffany L. Roe
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Scott D. Speer
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Hong Ji
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - LeeAnn Machiesky
- Process and Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Tianhui Zhang
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Deidre Wilkins
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Kevin M. Tuffy
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Elizabeth J. Kelly
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| |
Collapse
|
38
|
Nelson CB, Brady BL, Richards M, Lew CR, Via WL, Greenberg M, Rizzo C. Optimal site of care for administration of extended half-life respiratory syncytial virus (RSV) antibodies to infants in the United States (US). Vaccine 2023; 41:5820-5824. [PMID: 37586957 DOI: 10.1016/j.vaccine.2023.06.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION New extended half-life antibodies for the single-dose prevention of medically attended (MA) respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI) have been developed for administration to all infants before or during their first RSV season. For infants born during the season, administration as soon as feasible after birth would provide optimal protection and minimize access disparities. The objective of this study was to assess the time from birth hospitalization discharge to the first outpatient visit (FOV) among US infants in order to determine optimal site of administration for the extended half-life antibody. MATERIAL AND METHODS This retrospective, observational, time-to-event analysis uses the Merative™ MarketScan® Commercial and Multi-State Medicaid Databases. Time to FOV is reported separately for the COVID-19 and recent pre-COVID-19 eras and for commercially insured and Medicaid infants. RESULTS Overall, 73.8 % of Medicaid infants had an FOV within 5 days as compared to 84.7 % of commercially insured infants. Estimates were higher during the COVID-19 era. Urban commercially insured infants had much higher FOV completion than their counterparts. Among Medicaid infants, urban Black and rural White infants were least likely to complete their FOV within 5 days of birth hospitalization discharge. DISCUSSION AND CONCLUSION FOV within 5 days after birth hospitalization discharge for Medicaid infants is substantially lower than that of commercially insured infants. Approximately 1 in 4 Medicaid infants and 1 in 8 infants with commercial insurance did not have an outpatient visit within 5 days of birth hospitalization discharge. For US infants born during the RSV season, administration of extended half-life RSV antibodies in the newborn nursery prior to discharge would ensure optimal uptake and minimize access disparities.
Collapse
|
39
|
O’Hagan S, Galway N, Shields MD, Mallett P, Groves HE. Review of the Safety, Efficacy and Tolerability of Palivizumab in the Prevention of Severe Respiratory Syncytial Virus (RSV) Disease. Drug Healthc Patient Saf 2023; 15:103-112. [PMID: 37720805 PMCID: PMC10503506 DOI: 10.2147/dhps.s348727] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023] Open
Abstract
Respiratory Syncytial Virus (RSV) is a major global cause of childhood morbidity and mortality. Palivizumab, a monoclonal antibody that provides passive immunity against RSV, is currently licensed for prophylactic use in specific "high-risk" populations, including congenital heart disease, bronchopulmonary dysplasia and prematurity. Available research suggests palivizumab use in these high-risk populations can lead to a reduction in RSV-related hospitalization. However, palivizumab has not been demonstrated to reduce mortality, adverse events or length of hospital stay related to RSV. In this article, we review the management of RSV, indications for palivizumab prophylaxis, the safety, cost-effectiveness and efficacy of this preventative medication, and emerging therapeutics that could revolutionize future prevention of this significant pathogen.
Collapse
Affiliation(s)
- Shaun O’Hagan
- Paediatric Infectious Diseases, Royal Belfast Hospital for Sick Children, Belfast, Northern Ireland
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, Northern Ireland
| | - Niamh Galway
- Paediatric Respiratory Medicine, Royal Belfast Hospital for Sick Children, Belfast, Northern Ireland
| | - Michael D Shields
- Paediatric Respiratory Medicine, Royal Belfast Hospital for Sick Children, Belfast, Northern Ireland
- Centre for Medical Education, Queen’s University Belfast School of Medicine, Belfast, Northern Ireland
| | - Peter Mallett
- Paediatric Infectious Diseases, Royal Belfast Hospital for Sick Children, Belfast, Northern Ireland
- Centre for Medical Education, Queen’s University Belfast School of Medicine, Belfast, Northern Ireland
| | - Helen E Groves
- Paediatric Infectious Diseases, Royal Belfast Hospital for Sick Children, Belfast, Northern Ireland
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, Northern Ireland
| |
Collapse
|
40
|
Attaianese F, Guiducci S, Trapani S, Barbati F, Lodi L, Indolfi G, Azzari C, Ricci S. Reshaping Our Knowledge: Advancements in Understanding the Immune Response to Human Respiratory Syncytial Virus. Pathogens 2023; 12:1118. [PMID: 37764926 PMCID: PMC10536346 DOI: 10.3390/pathogens12091118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) is a significant cause of respiratory tract infections, particularly in young children and older adults. In this review, we aimed to comprehensively summarize what is known about the immune response to hRSV infection. We described the innate and adaptive immune components involved, including the recognition of RSV, the inflammatory response, the role of natural killer (NK) cells, antigen presentation, T cell response, and antibody production. Understanding the complex immune response to hRSV infection is crucial for developing effective interventions against this significant respiratory pathogen. Further investigations into the immune memory generated by hRSV infection and the development of strategies to enhance immune responses may hold promise for the prevention and management of hRSV-associated diseases.
Collapse
Affiliation(s)
- Federica Attaianese
- Postgraduate School of Pediatrics, University of Florence, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy;
| | - Sara Guiducci
- Postgraduate School of Immunology, University of Florence, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy;
| | - Sandra Trapani
- Pediatric Unit, Meyer Children’s Hospital IRCCS, Viale Pieraccini 24, 50139 Florence, Italy; (S.T.); (G.I.)
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (L.L.); (C.A.)
| | - Federica Barbati
- Postgraduate School of Pediatrics, University of Florence, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy;
| | - Lorenzo Lodi
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (L.L.); (C.A.)
- Division of Immunology, Meyer Children’s Hospital IRCCS, Viale Pieraccini 24, 50139 Florence, Italy
| | - Giuseppe Indolfi
- Pediatric Unit, Meyer Children’s Hospital IRCCS, Viale Pieraccini 24, 50139 Florence, Italy; (S.T.); (G.I.)
- NEUROFARBA Department, University of Florence, 50139 Florence, Italy
| | - Chiara Azzari
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (L.L.); (C.A.)
- Division of Immunology, Meyer Children’s Hospital IRCCS, Viale Pieraccini 24, 50139 Florence, Italy
| | - Silvia Ricci
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (L.L.); (C.A.)
- Division of Immunology, Meyer Children’s Hospital IRCCS, Viale Pieraccini 24, 50139 Florence, Italy
| |
Collapse
|
41
|
Bracht M, Rodgers-Gray B, Bacchini F, Paes BA. Understanding Policy Decisions and Their Implications Regarding Preventive Interventions for Respiratory Syncytial Virus (RSV) Infection in Canadian Infants: A Primer for Nurses. Neonatal Netw 2023; 42:291-302. [PMID: 37657806 DOI: 10.1891/nn-2023-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 09/03/2023]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of morbidity and hospitalization in young children, and prevention is the primary management strategy. At present, palivizumab, a monoclonal antibody providing immediate passive immunity, rather than a vaccine that induces active immunity, is the only preventive intervention used in routine practice internationally. In Canada, access varies across the country. Prophylaxis policies are mainly driven by cost-effectiveness analyses, and it is crucial that the full costs and benefits of any intervention are captured. Positive results from a new Canadian cost-effectiveness analysis of palivizumab will help address the current inequality in use while providing a framework for future models of RSV preventives. Nurses are the principal educators for parents about the risks of childhood RSV and optimal prevention via basic hygiene, behavioral and environmental measures, and seasonal prophylaxis. Nurses should be provided not only with regular, up-to-date, and accurate information on RSV and the clinical aspects of emerging interventions but be informed on the decision-making governing the use of preventive strategies.
Collapse
|
42
|
Ahani B, Tuffy KM, Aksyuk AA, Wilkins D, Abram ME, Dagan R, Domachowske JB, Guest JD, Ji H, Kushnir A, Leach A, Madhi SA, Mankad VS, Simões EAF, Sparklin B, Speer SD, Stanley AM, Tabor DE, Hamrén UW, Kelly EJ, Villafana T. Molecular and phenotypic characteristics of RSV infections in infants during two nirsevimab randomized clinical trials. Nat Commun 2023; 14:4347. [PMID: 37468530 PMCID: PMC10356750 DOI: 10.1038/s41467-023-40057-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Nirsevimab is a monoclonal antibody that binds to the respiratory syncytial virus (RSV) fusion protein. During the Phase 2b (NCT02878330) and MELODY (NCT03979313) clinical trials, infants received one dose of nirsevimab or placebo before their first RSV season. In this pre-specified analysis, isolates from RSV infections were subtyped, sequenced and analyzed for nirsevimab binding site substitutions; subsequently, recombinant RSVs were engineered for microneutralization susceptibility testing. Here we show that the frequency of infections caused by subtypes A and B is similar across and within the two trials. In addition, RSV A had one and RSV B had 10 fusion protein substitutions occurring at >5% frequency. Notably, RSV B binding site substitutions were rare, except for the highly prevalent I206M:Q209R, which increases nirsevimab susceptibility; RSV B isolates from two participants had binding site substitutions that reduce nirsevimab susceptibility. Overall, >99% of isolates from the Phase 2b and MELODY trials retained susceptibility to nirsevimab.
Collapse
Affiliation(s)
- Bahar Ahani
- Bioinformatics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kevin M Tuffy
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anastasia A Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Michael E Abram
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ron Dagan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences of the Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Johnathan D Guest
- Virology and Vaccine Discovery, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Hong Ji
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anna Kushnir
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vaishali S Mankad
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - Eric A F Simões
- University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Benjamin Sparklin
- Bioinformatics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Scott D Speer
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - David E Tabor
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
43
|
Antillón M, Li X, Willem L, Bilcke J, RESCEU investigators, Jit M, Beutels P. The age profile of respiratory syncytial virus burden in preschool children of low- and middle-income countries: A semi-parametric, meta-regression approach. PLoS Med 2023; 20:e1004250. [PMID: 37459352 PMCID: PMC10389726 DOI: 10.1371/journal.pmed.1004250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 07/31/2023] [Accepted: 05/30/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infections are among the primary causes of death for children under 5 years of age worldwide. A notable challenge with many of the upcoming prophylactic interventions against RSV is their short duration of protection, making the age profile of key interest to the design of prevention strategies. METHODS AND FINDINGS We leverage the RSV data collected on cases, hospitalizations, and deaths in a systematic review in combination with flexible generalized additive mixed models (GAMMs) to characterize the age burden of RSV incidence, hospitalization, and hospital-based case fatality rate (hCFR). Due to the flexible nature of GAMMs, we estimate the peak, median, and mean incidence of infection to inform discussions on the ideal "window of protection" of prophylactic interventions. In a secondary analysis, we reestimate the burden of RSV in all low- and middle-income countries. The peak age of community-based incidence is 4.8 months, and the mean and median age of infection is 18.9 and 14.7 months, respectively. Estimating the age profile using the incidence coming from hospital-based studies yields a slightly younger age profile, in which the peak age of infection is 2.6 months and the mean and median age of infection are 15.8 and 11.6 months, respectively. More severe outcomes, such as hospitalization and in-hospital death have a younger age profile. Children under 6 months of age constitute 10% of the population under 5 years of age but bear 20% to 29% of cases, 28% to 39% of hospitalizations, and 38% to 50% of deaths. On an average year, we estimate 28.23 to 31.34 million cases of RSV, between 2.95 to 3.35 million hospitalizations, and 16,835 to 19,909 in-hospital deaths in low, lower- and upper middle-income countries. In addition, we estimate 17,254 to 23,875 deaths in the community, for a total of 34,114 to 46,485 deaths. Globally, evidence shows that community-based incidence may differ by World Bank Income Group, but not hospital-based incidence, probability of hospitalization, or the probability of in-hospital death (p ≤ 0.01, p = 1, p = 0.86, 0.63, respectively). Our study is limited mainly due to the sparsity of the data, especially for low-income countries (LICs). The lack of information for some populations makes detecting heterogeneity between income groups difficult, and differences in access to care may impact the reported burden. CONCLUSIONS We have demonstrated an approach to synthesize information on RSV outcomes in a statistically principled manner, and we estimate that the age profile of RSV burden depends on whether information on incidence is collected in hospitals or in the community. Our results suggest that the ideal prophylactic strategy may require multiple products to avert the risk among preschool children.
Collapse
Affiliation(s)
- Marina Antillón
- Center for Health Economics and Modeling of Infectious Diseases, University of Antwerp, Antwerp, Belgium
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Xiao Li
- Center for Health Economics and Modeling of Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Lander Willem
- Center for Health Economics and Modeling of Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Joke Bilcke
- Center for Health Economics and Modeling of Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | | | - Mark Jit
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Philippe Beutels
- Center for Health Economics and Modeling of Infectious Diseases, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
44
|
Esposito S, Amirthalingam G, Bassetti M, Blasi F, De Rosa FG, Halasa NB, Hung I, Osterhaus A, Tan T, Torres JP, Vena A, Principi N. Monoclonal antibodies for prophylaxis and therapy of respiratory syncytial virus, SARS-CoV-2, human immunodeficiency virus, rabies and bacterial infections: an update from the World Association of Infectious Diseases and Immunological Disorders and the Italian Society of Antinfective Therapy. Front Immunol 2023; 14:1162342. [PMID: 37256125 PMCID: PMC10226646 DOI: 10.3389/fimmu.2023.1162342] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
Monoclonal antibodies (mABs) are safe and effective proteins produced in laboratory that may be used to target a single epitope of a highly conserved protein of a virus or a bacterial pathogen. For this purpose, the epitope is selected among those that play the major role as targets for prevention of infection or tissue damage. In this paper, characteristics of the most important mABs that have been licensed and used or are in advanced stages of development for use in prophylaxis and therapy of infectious diseases are discussed. We showed that a great number of mABs effective against virus or bacterial infections have been developed, although only in a small number of cases these are licensed for use in clinical practice and have reached the market. Although some examples of therapeutic efficacy have been shown, not unlike more traditional antiviral or antibacterial treatments, their efficacy is significantly greater in prophylaxis or early post-exposure treatment. Although in many cases the use of vaccines is more effective and cost-effective than that of mABs, for many infectious diseases no vaccines have yet been developed and licensed. Furthermore, in emergency situations, like in epidemics or pandemics, the availability of mABs can be an attractive adjunct to our armament to reduce the impact. Finally, the availability of mABs against bacteria can be an important alternative, when multidrug-resistant strains are involved.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gayatri Amirthalingam
- Immunisation and Countermeasures Division, National Infection Service, Public Health England, London, United Kingdom
| | - Matteo Bassetti
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | | | - Natasha B. Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ivan Hung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Foundation, Hannover, Germany
| | - Tina Tan
- Division of Infectious Diseases, Feinberg School of Medicine of Northwestern University, Chicago, IL, United States
| | - Juan Pablo Torres
- Department of Pediatrics and Pediatric Surgery, Facultad de Medicina, University of Chile, Santiago, Chile
- Instituto Sistemas Complejos de Ingeniería (ISCI), Santiago, Chile
| | - Antonio Vena
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
45
|
Wilkins D, Yuan Y, Chang Y, Aksyuk AA, Núñez BS, Wählby-Hamrén U, Zhang T, Abram ME, Leach A, Villafana T, Esser MT. Durability of neutralizing RSV antibodies following nirsevimab administration and elicitation of the natural immune response to RSV infection in infants. Nat Med 2023; 29:1172-1179. [PMID: 37095249 PMCID: PMC10202809 DOI: 10.1038/s41591-023-02316-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Nirsevimab is an extended half-life monoclonal antibody specific for the prefusion conformation of the respiratory syncytial virus (RSV) F protein, which has been studied in preterm and full-term infants in the phase 2b and phase 3 MELODY trials. We analyzed serum samples collected from 2,143 infants during these studies to characterize baseline levels of RSV-specific immunoglobulin G antibodies and neutralizing antibodies (NAbs), duration of RSV NAb levels following nirsevimab administration, the risk of RSV exposure during the first year of life and the infant's adaptive immune response to RSV following nirsevimab administration. Baseline RSV antibody levels varied widely; consistent with reports that maternal antibodies are transferred late in the third trimester, preterm infants had lower baseline RSV antibody levels than full-term infants. Nirsevimab recipients had RSV NAb levels >140-fold higher than baseline at day 31 and remained >50-fold higher at day 151 and >7-fold higher at day 361. Similar seroresponse rates to the postfusion form of RSV F protein in nirsevimab recipients (68-69%) compared with placebo recipients (63-70%; not statistically significant) suggest that while nirsevimab protects from RSV disease, it still allows an active immune response. In summary, nirsevimab provided sustained, high levels of NAb throughout an infant's first RSV season and prevented RSV disease while allowing the development of an immune response to RSV.
Collapse
Affiliation(s)
- Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Yuan Yuan
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yue Chang
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anastasia A Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Beatriz Seoane Núñez
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Madrid, Spain
| | - Ulrika Wählby-Hamrén
- Clinical Pharmacology & Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tianhui Zhang
- Data Sciences and Quantitative Biology, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Michael E Abram
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Mark T Esser
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
46
|
Convertino I, Ferraro S, Cappello E, Valdiserra G, Bonaso M, Tuccori M. Tixagevimab + cilgavimab against SARS-CoV-2: the preclinical and clinical development and real-world evidence. Expert Opin Drug Discov 2023; 18:231-245. [PMID: 36649625 DOI: 10.1080/17460441.2023.2170348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Direct-acting SARS-CoV-2 antiviral monoclonal antibodies have been an integral part of therapeutic strategies against COVID-19 pandemic. The monoclonal strategy was jeopardized by the emergence of new variants and resistant strains, making many monoclonal antibodies quickly obsolete. Nevertheless, a possible strategy consists in the use of antibody cocktails and the development of the cilgavimab + tixagevimab in combination is placed in this context. AREAS COVERED In this review, we describe the development of the cilgavimab + tixagevimab cocktail, from pre-clinical to real-world evidence. EXPERT OPINION The pre-clinical and clinical development of cilgavimab + tixagevimab followed a similar path to that of the antibodies developed in the earlier stages of the pandemic. Both antibodies have been developed from convalescent plasma and have been shown to be effective in clinical trials in prophylaxis and in early therapy. This cocktail has found its position in therapy especially in immunocompromised subjects for whom vaccine prevention is not feasible. The cocktail strategy, together with a more stable pandemic situation, could ensure a certain longevity to the drug against resistance, especially when compared with that of other antibodies. Recently emerged Omicron sub-lineages have demonstrated the ability to escape this cocktail's activity and so the future of this treatment could be compromised.
Collapse
Affiliation(s)
- Irma Convertino
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sara Ferraro
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Emiliano Cappello
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giulia Valdiserra
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco Bonaso
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco Tuccori
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Unit of Adverse Drug Reactions Monitoring, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
47
|
Simões EAF, Madhi SA, Muller WJ, Atanasova V, Bosheva M, Cabañas F, Baca Cots M, Domachowske JB, Garcia-Garcia ML, Grantina I, Nguyen KA, Zar HJ, Berglind A, Cummings C, Griffin MP, Takas T, Yuan Y, Wählby Hamrén U, Leach A, Villafana T. Efficacy of nirsevimab against respiratory syncytial virus lower respiratory tract infections in preterm and term infants, and pharmacokinetic extrapolation to infants with congenital heart disease and chronic lung disease: a pooled analysis of randomised controlled trials. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:180-189. [PMID: 36634694 PMCID: PMC9940918 DOI: 10.1016/s2352-4642(22)00321-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND In a phase 2b trial and the phase 3 MELODY trial, nirsevimab, an extended half-life, monoclonal antibody against respiratory syncytial virus (RSV), protected healthy infants born preterm or at full term against medically attended RSV lower respiratory tract infection (LRTI). In the MEDLEY phase 2-3 trial in infants at higher risk for severe RSV infection, nirsevimab showed a similar safety profile to that of palivizumab. The aim of the current analysis was to assess the efficacy of nirsevimab using a weight-banded dosing regimen in infants born between 29 weeks gestational age and full term. METHODS Infants enrolled in the phase 2b and MELODY trials were randomised (2:1) to receive a single intramuscular injection of nirsevimab (infants weighing <5 kg received 50 mg; those weighing ≥5 kg received 100 mg) or placebo before the RSV season. Infants in MEDLEY were randomised (2:1) to receive one dose of nirsevimab (infants weighing <5 kg received 50 mg; those weighing ≥5 kg received 100 mg) followed by four monthly placebo doses, or five once-a-month intramuscular doses of palivizumab. We report a prespecified pooled efficacy analysis assessing the weight-banded dosing regimen proposed on the basis of the phase 2b and MELODY trials, in addition to extrapolated efficacy in infants with chronic lung disease, congenital heart disease, or extreme preterm birth (<29 weeks' gestational age) based on pharmacokinetic data from the phase 2-3 MEDLEY safety trial. For the pooled efficacy analysis, the primary endpoint was incidence of medically attended RSV LRTI through 150 days post-dose. The secondary efficacy endpoint was number of admissions to hospital for medically attended RSV LRTI. The incidence of very severe RSV LRTI was an exploratory endpoint, defined as cases of hospital admission for medically attended RSV LRTI that required supplemental oxygen or intravenous fluids. We also did a prespecified exploratory analysis of medically attended LRTI of any cause (in the investigator's judgement) and hospital admission for respiratory illness of any cause (defined as any upper respiratory tract infection or LRTI leading to hospital admission). Post hoc exploratory analyses of outpatient visits and antibiotic use were also done. Nirsevimab serum concentrations in MEDLEY were assessed using population pharmacokinetic methods and the pooled data from the phase 2b and MELODY trials. An exposure target was defined on the basis of an exposure-response analysis. To successfully demonstrate extrapolation, more than 80% of infants in MEDLEY had to achieve serum nirsevimab exposures at or above the predicted efficacious target. FINDINGS Overall, 2350 infants (1564 in the nirsevimab group and 786 in the placebo group) in the phase 2b and MELODY trials were included in the pooled analysis. Nirsevimab showed efficacy versus placebo with respect to the primary endpoint of medically attended RSV LRTI (19 [1%] nirsevimab recipients vs 51 [6%] placebo recipients; relative risk reduction [RRR] 79·5% [95% CI 65·9-87·7]). Consistent efficacy was shown for additional endpoints of RSV LRTI hospital admission (nine [1%] nirsevimab recipients vs 21 [3%] placebo recipients; 77·3% [50·3-89·7]) and very severe RSV (five [<1%] vs 18 [2%]; 86·0% [62·5-94·8]). Nirsevimab recipients had fewer hospital admissions for any-cause respiratory illness (RRR 43·8% [18·8-61·1]), any-cause medically attended LRTI (35·4% [21·5-46·9]), LRTI outpatient visits (41·9% [25·7-54·6]), and antibiotic prescriptions (23·6% [3·8-39·3]). Among infants with chronic lung disease, congenital heart disease, or extreme preterm birth in MEDLEY, nirsevimab serum exposures were similar to those found in the pooled data; exposures were above the target in more than 80% of the overall MEDLEY trial population (94%), including infants with chronic lung disease (94%) or congenital heart disease (80%) and those born extremely preterm (94%). INTERPRETATION A single dose of nirsevimab protected healthy infants born at term or preterm from medically attended RSV LRTI, associated hospital admission, and severe RSV. Pharmacokinetic data support efficacy extrapolation to infants with chronic lung disease, congenital heart disease, or extreme prematurity. Together, these data suggest that nirsevimab has the potential to change the landscape of infant RSV disease by reducing a major cause of infant morbidity and the consequent burden on caregivers, clinicians, and health-care providers. FUNDING AstraZeneca and Sanofi.
Collapse
Affiliation(s)
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - William J Muller
- Ann & Robert H Lurie Children's Hospital of Chicago and Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Miroslava Bosheva
- University Multiprofile Hospital for Active Treatment Sv Georgi Medical University, Plovdiv, Bulgaria
| | | | | | | | | | | | - Kim A Nguyen
- Hospices Civils de Lyon, Neonatal Intensive Care Units and CIC 1407, Lyon, France
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross Children's Hospital, Cape Town, South Africa; SA-MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Anna Berglind
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Celeste Cummings
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - M Pamela Griffin
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Therese Takas
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yuan Yuan
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
48
|
Do LAH, Vodicka E, Nguyen A, Le TNK, Nguyen TTH, Thai QT, Pham VQ, Pham TU, Nguyen TN, Mulholland K, Cao MT, Le NTN, Tran AT, Pecenka C. Estimating the economic burden of respiratory syncytial virus infections in infants in Vietnam: a cohort study. BMC Infect Dis 2023; 23:73. [PMID: 36747128 PMCID: PMC9901829 DOI: 10.1186/s12879-023-08024-2] [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: 09/08/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Little information is available on the costs of respiratory syncytial virus (RSV) in Vietnam or other low- and middle-income countries. Our study estimated the costs of LRTIs associated with RSV infection among children in southern Vietnam. METHODS We conducted a prospective cohort study evaluating household and societal costs associated with LRTIs stratified by RSV status and severity among children under 2 years old who sought care at a major pediatric referral hospital in southern Vietnam. Enrollment periods were September 2019-December 2019, October 2020-June 2021 and October 2021-December 2021. RSV status was confirmed by a validated RT-PCR assay. RSV rapid detection antigen (RDA) test performance was also evaluated. Data on resource utilization, direct medical and non-medical costs, and indirect costs were collected from billing records and supplemented by patient-level questionnaires. All costs are reported in 2022 US dollars. RESULTS 536 children were enrolled in the study, with a median age of 7 months (interquartile range [IQR] 3-12). This included 210 (39.2%) children from the outpatient department, 318 children (59.3%) from the inpatient respiratory department (RD), and 8 children (1.5%) from the intensive care unit (ICU). Nearly 20% (105/536) were RSV positive: 3.9 percent (21/536) from the outpatient department, 15.7% (84/536) from the RD, and none from the ICU. The median total cost associated with LRTI per patient was US$52 (IQR 32-86) for outpatients and US$184 (IQR 109-287) for RD inpatients. For RSV-associated LRTIs, the median total cost per infection episode per patient was US$52 (IQR 32-85) for outpatients and US$165 (IQR 95-249) for RD inpatients. Total out-of-pocket costs of one non-ICU admission of RSV-associated LRTI ranged from 32%-70% of the monthly minimum wage per person (US$160) in Ho Chi Minh City. The sensitivity and the specificity of RSV RDA test were 88.2% (95% CI 63.6-98.5%) and 100% (95% CI 93.3-100%), respectively. CONCLUSION These are the first data reporting the substantial economic burden of RSV-associated illness in young children in Vietnam. This study informs policymakers in planning health care resources and highlights the urgency of RSV disease prevention.
Collapse
Affiliation(s)
- Lien Anh Ha Do
- grid.1058.c0000 0000 9442 535XNew Vaccine Group, Murdoch Children’s Research Institute, 50 Flemington Road, Parkville, Melbourne, 3051 Australia ,grid.1008.90000 0001 2179 088XDepartment of Pediatrics, The University of Melbourne, Melbourne, Australia
| | | | | | - Thi Ngoc Kim Le
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | - Thi Thanh Hai Nguyen
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | - Quang Tung Thai
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | - Van Quang Pham
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | - Thanh Uyen Pham
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | - Thu Ngoc Nguyen
- grid.452689.4Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Kim Mulholland
- grid.1058.c0000 0000 9442 535XNew Vaccine Group, Murdoch Children’s Research Institute, 50 Flemington Road, Parkville, Melbourne, 3051 Australia ,grid.1008.90000 0001 2179 088XDepartment of Pediatrics, The University of Melbourne, Melbourne, Australia ,grid.8991.90000 0004 0425 469XLondon School of Hygiene and Tropical Medicine, London, UK
| | - Minh Thang Cao
- grid.452689.4Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Nguyen Thanh Nhan Le
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | - Anh Tuan Tran
- grid.440249.f0000 0004 4691 4406Children’s Hospital 1, Ho Chi Minh City, Vietnam
| | | |
Collapse
|
49
|
Sun M, Lai H, Na F, Li S, Qiu X, Tian J, Zhang Z, Ge L. Monoclonal Antibody for the Prevention of Respiratory Syncytial Virus in Infants and Children: A Systematic Review and Network Meta-analysis. JAMA Netw Open 2023; 6:e230023. [PMID: 36800182 PMCID: PMC9938429 DOI: 10.1001/jamanetworkopen.2023.0023] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
IMPORTANCE Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory infection in children younger than 5 years; effective prevention strategies are urgently needed. OBJECTIVE To compare the efficacy and safety of monoclonal antibodies for the prevention of RSV infection in infants and children. DATA SOURCES In this systematic review and network meta-analysis, PubMed, Embase, CENTRAL, and ClinicalTrials.gov were searched from database inception to March 2022. STUDY SELECTION Randomized clinical trials that enrolled infants at high risk of RSV infection to receive a monoclonal antibody or placebo were included. Keywords and extensive vocabulary related to monoclonal antibodies, RSV, and randomized clinical trials were searched. DATA EXTRACTION AND SYNTHESIS The Preferred Reporting Items for Systematic Reviews and Meta-analyses reporting guideline was used. Teams of 2 reviewers independently performed literature screening, data extraction, and risk of bias assessment. The Grading of Recommendations, Assessments, Developments, and Evaluation approach was used to rate the certainty of evidence. A random-effects model network meta-analysis was conducted using a consistency model under the frequentist framework. MAIN OUTCOMES AND MEASURES The main outcomes were all-cause mortality, RSV-related hospitalization, RSV-related infection, drug-related adverse events, intensive care unit admission, supplemental oxygen use, and mechanical ventilation use. RESULTS Fifteen randomized clinical trials involving 18 395 participants were eligible; 14 were synthesized, with 18 042 total participants (median age at study entry, 3.99 months [IQR, 3.25-6.58 months]; median proportion of males, 52.37% [IQR, 50.49%-53.85%]). Compared with placebo, with moderate- to high-certainty evidence, nirsevimab, palivizumab, and motavizumab were associated with significantly reduced RSV-related infections per 1000 participants (nirsevimab: -123 [95% CI, -138 to -100]; palivizumab: -108 [95% CI, -127 to -82]; motavizumab: -136 [95% CI, -146 to -125]) and RSV-related hospitalizations per 1000 participants (nirsevimab: -54 [95% CI, -64 to -38; palivizumab: -39 [95% CI, -48 to -28]; motavizumab: -48 [95% CI, -58 to -33]). With moderate-certainty evidence, both motavizumab and palivizumab were associated with significant reductions in intensive care unit admissions per 1000 participants (-8 [95% CI, -9 to -4] and -5 [95% CI, -7 to 0], respectively) and supplemental oxygen use per 1000 participants (-59 [95% CI, -63 to -54] and -55 [95% CI, -61 to -41], respectively), and nirsevimab was associated with significantly reduced supplemental oxygen use per 1000 participants (-59 [95% CI, -65 to -40]). No significant differences were found in all-cause mortality and drug-related adverse events. Suptavumab did not show any significant benefits for the outcomes of interest. CONCLUSIONS AND RELEVANCE In this study, motavizumab, nirsevimab, and palivizumab were associated with substantial benefits in the prevention of RSV infection, without a significant increase in adverse events compared with placebo. However, more research is needed to confirm the present conclusions, especially for safety and cost-effectiveness.
Collapse
Affiliation(s)
- Mingyao Sun
- Department of Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
- Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Honghao Lai
- Evidence-Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
- Department of Social Medicine and Health Management, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Feiyang Na
- Department of Pediatrics, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu, China
| | - Sheng Li
- The First People’s Hospital of Lanzhou City, Lanzhou, Gansu, China
| | - Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Chengdu, Sichuan, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Zhigang Zhang
- Department of Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
- Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Long Ge
- Evidence-Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
- Department of Social Medicine and Health Management, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
50
|
Raes M, Daelemans S, Cornette L, Moniotte S, Proesmans M, Schaballie H, Frère J, Vanden Driessche K, Van Brusselen D. The burden and surveillance of RSV disease in young children in Belgium-expert opinion. Eur J Pediatr 2023; 182:451-460. [PMID: 36371521 PMCID: PMC9660201 DOI: 10.1007/s00431-022-04698-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
UNLABELLED Infections with respiratory syncytial virus (RSV) can cause severe disease. In young children, RSV is the most common cause of lower respiratory tract illness and life-threatening infections most commonly occur in the first years of life. In adults, elderly and immunocompromised people are most vulnerable. Recently there has been an acceleration in the development of candidate RSV vaccines, monoclonal antibodies and therapeutics which are expected to become available in Europe within the next 2-10 years. Understanding the true burden of childhood RSV disease will become very important to support public health authorities and policy makers in the assessment of new therapeutic opportunities against RSV disease. A systematic literature search was performed to map local data on the burden of RSV disease and to evaluate available RSV surveillance systems. A group of 9 paediatric infectious diseases specialists participated in an expert panel. The purpose of this meeting was to evaluate and map the burden associated with RSV infection in children, including patient pathways and the epidemiological patterns of virus circulation in Belgium. Sources of information on the burden of RSV disease in Belgium are very limited. For the outpatient setting, it is estimated that 5-10% of young patients seen in primary care are referred to the hospital. Around 3500 children between 0 and 12 months of age are hospitalized for RSV-bronchiolitis every year and represent the majority of all hospitalizations. The current Belgian RSV surveillance system was evaluated and found to be insufficient. Knowledge gaps are highlighted and future perspectives and priorities offered. CONCLUSION The Belgian population-based RSV surveillance should be improved, and a hospital-led reporting system should be put in place to enable the evaluation of the true burden of RSV disease in Belgium and to improve disease management in the future. WHAT IS KNOWN • RSV bronchiolitis is a very important cause of infant hospitalization. • The burden of disease in the community is poorly studied and underestimated. WHAT IS NEW • This expert opinion summarizes knowledge gaps and offers insights that allow improvement of local surveillance systems in order to establish a future-proof RSV surveillance system.
Collapse
Affiliation(s)
- Marc Raes
- Department of Paediatrics, Jessa Hospital, Hasselt, Belgium.
| | - Siel Daelemans
- Paediatric Pulmonary and Infectious Diseases, University Hospital Brussel, Brussels, Belgium
| | - Luc Cornette
- Department of Neonatology, AZ Sint-Jan Hospital, Brugge, Belgium
| | - Stéphane Moniotte
- Department of Paediatric Cardiology, University Hospital Saint-Luc, UCLouvain, Brussels, Belgium
| | - Marijke Proesmans
- Paediatric Department, University Hospital Gasthuisberg, Leuven, Belgium
| | - Heidi Schaballie
- Department of Paediatric Pulmonology, Infectious Diseases and Immune Disorders, University Hospital, Ghent, Belgium
| | - Julie Frère
- Department of Paediatrics and Infectious Diseases, University Hospital, Liège, Belgium
| | | | - Daan Van Brusselen
- Department of Paediatric Infectious Diseases, GZA Hospitals, Antwerp, Belgium
| |
Collapse
|