1
|
Zhou J, Rao R, Shapiro ME, Tania N, Herron C, Musante CJ, Hughes JH. Model-Informed Drug Development Applications and Opportunities in mRNA-LNP Therapeutics. Clin Pharmacol Ther 2025. [PMID: 40083288 DOI: 10.1002/cpt.3641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
The utilization of lipid nanoparticles (LNP) for encapsulating mRNA has revolutionized the field of therapeutics, enabling the rapid development of COVID-19 vaccines and cancer vaccines. However, the clinical development of mRNA-LNP therapeutics faces numerous challenges due to their complex mechanisms of action and limited clinical experience. To overcome these hurdles, Model-Informed Drug Development (MIDD) emerges as a valuable tool that can be applied to mRNA-LNP therapeutics, facilitating the evaluation of their safety and efficacy through the integration of data from all stages into appropriate modeling and simulation techniques. In this review, we provide an overview of current MIDD applications in mRNA-LNP therapeutics clinical development using in vivo data. A variety of modeling methods are reviewed, including quantitative system pharmacology (QSP), physiologically based pharmacokinetics (PBPK), mechanistic pharmacokinetics/pharmacodynamics (PK/PD), population PK/PD, and model-based meta-analysis (MBMA). Additionally, we compare the differences between mRNA-based therapeutics, small interfering RNA, and adeno-associated virus-based gene therapies in terms of their clinical pharmacology, and discuss the potential for mutual sharing of MIDD knowledge between these therapeutics. Furthermore, we highlight the promising future opportunities for applying MIDD approaches in the development of mRNA-LNP drugs. By emphasizing the importance of applying MIDD knowledge throughout mRNA-LNP therapeutics development, this review aims to encourage stakeholders to recognize the value of MIDD and its potential to enhance the safety and efficacy evaluation of mRNA-LNP therapeutics.
Collapse
Affiliation(s)
| | - Rohit Rao
- Pfizer Inc, Cambridge, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
2
|
Dari A, Pérez Ruixo JJ, Le Gars M, Struyf F, Jacqmin P. Modelling antibody dynamics in humans after different Ad26.COV2.S vaccination schemes. Br J Clin Pharmacol 2025; 91:397-408. [PMID: 39327825 DOI: 10.1111/bcp.16251] [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: 03/18/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 09/28/2024] Open
Abstract
AIMS To develop a semimechanistic model that describes the kinetic profile and variability of antibody (Ab) concentrations following vaccination with Ad26.COV2.S at different doses and dosing intervals. METHODS Data were collected from participants randomized into 5 clinical trials receiving the Ad26.COV2.S vaccine. The model considered key elements of humoral immune response, dose proportionality and the evolutionary processes of the immune response. Interindividual variability and covariates were explored. RESULTS Fast and slow kinetic phases of Ab and their evolution over time were differentiated. After first and second administrations, Ab concentrations of both phases increased less than dose proportionally, indicating a saturation of B-cell production processes. Ab concentrations produced during the fast kinetic phase increased significantly after the second administration, indicating an underlying evolutive process after antigen exposures. For the slow kinetic phase, a less pronounced increase occurred after the second and third administrations but was relatively higher in subjects who had low concentrations after the first administration. Ab concentrations of the slow phase were higher in females and decreased with age. After multiple administrations, the fast phase had Ab maximum concentrations about 5 times higher than the slow phase. The limiting kinetic factors in the fast and slow phases were the elimination rates of Ab itself and Ab producing cells, respectively. CONCLUSION The model appears suitable to quantitatively describe the inter- and intraindividual kinetics of the immune response and the impact of covariates after multiple administrations of a vaccine.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research & Development, Beerse, Belgium
| | | | | | | | | |
Collapse
|
3
|
Süner A, Ergör G, Çağlayan D, Türe N, Güzel I, Irmak Ç, Işık E, Appak Ö, Çelik M, Öztürk G, Çavuş S, Sayiner A, Ergör A, Demiral Y, Kilic B. Waning Humoral Immune Response Following the Third and Fourth SARS-COV-2 Vaccine: A Cohort Study in Healthcare Workers. Influenza Other Respir Viruses 2024; 18:e70003. [PMID: 39189319 PMCID: PMC11347934 DOI: 10.1111/irv.70003] [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: 04/26/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND This study is aimed at providing information about the timing of booster doses and antibody kinetics in healthcare workers. METHODS This research extends a prospective cohort study conducted at Dokuz Eylul University Hospital in Turkey, covering the period from March 2021 to December 2021. During this timeframe, the antibody levels of the health workers were measured at four different time points. The associations of antibody levels with gender, age, occupation, body mass index (BMI), chronic disease, and smoking were analyzed. RESULTS There was a significant difference between antibody levels in all four blood draws (p < 0.001). Antibody levels decreased in both those vaccinated with BNT162b2 (p < 0.001) and those vaccinated with CoronaVac (p = 0.002) until the fourth blood draw. There was a significant difference between those vaccinated with one and two doses of booster BNT162b2 before the third blood draw (p < 0.001), which continued at the fourth blood draw (p < 0.001). The antibody levels of those with an interval of 41-50 days between two vaccinations decreased significantly at the fourth blood draw (p < 0.001). CONCLUSIONS This study provides insight into the dynamics and persistence of antibody response after additional COVID-19 vaccine doses among healthcare workers. The longer the interval between booster doses may result in greater antibody levels being maintained over time, allowing for longer durations of protection.
Collapse
Affiliation(s)
- Ahmet Furkan Süner
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Gül Ergör
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Derya Çağlayan
- Infectious Diseases UnitDiyarbakır Provincial Health DirectorateDiyarbakirTurkey
| | - Neslişah Türe
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Irmak Güzel
- Medical Microbiology UnitMardin Nusaybin State HospitalMardinTurkey
| | - Çağlar Irmak
- Infectious Diseases and Clinical Microbiology UnitHakkari Yüksekova State HospitalHakkariTurkey
| | - Elif Işık
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Özgür Appak
- Department of Medical Microbiology, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Muammer Çelik
- Department of Infectious Diseases and Clinical Microbiology, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Gamze Öztürk
- Department of Medical Microbiology, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Sema Alp Çavuş
- Department of Infectious Diseases and Clinical Microbiology, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Arzu Sayiner
- Department of Medical Microbiology, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Alp Ergör
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Yücel Demiral
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| | - Bulent Kilic
- Department of Public Health, Faculty of MedicineDokuz Eylul UniversityIzmirTurkey
| |
Collapse
|
4
|
Sumi T, Harada K. Vaccine and antiviral drug promise for preventing post-acute sequelae of COVID-19, and their combination for its treatment. Front Immunol 2024; 15:1329162. [PMID: 39185419 PMCID: PMC11341427 DOI: 10.3389/fimmu.2024.1329162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Most healthy individuals recover from acute SARS-CoV-2 infection, whereas a remarkable number continues to suffer from unexplained symptoms, known as Long COVID or post-acute COVID-19 syndrome (PACS). It is therefore imperative that methods for preventing and treating the onset of PASC be investigated with the utmost urgency. Methods A mathematical model of the immune response to vaccination and viral infection with SARS-CoV-2, incorporating immune memory cells, was developed. Results and discussion Similar to our previous model, persistent infection was observed by the residual virus in the host, implying the possibility of chronic inflammation and delayed recovery from tissue injury. Pre-infectious vaccination and antiviral medication administered during onset can reduce the acute viral load; however, they show no beneficial effects in preventing persistent infection. Therefore, the impact of these treatments on the PASC, which has been clinically observed, is mainly attributed to their role in preventing severe tissue damage caused by acute viral infections. For PASC patients with persistent infection, vaccination was observed to cause an immediate rapid increase in viral load, followed by a temporary decrease over approximately one year. The former was effectively suppressed by the coadministration of antiviral medications, indicating that this combination is a promising treatment for PASC.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, Okayama, Japan
- Department of Chemistry, Faculty of Science, Okayama University, Okayama, Japan
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, Toyohashi, Aichi, Japan
- Center for IT-Based Education, Toyohashi University of Technology, Toyohashi, Aichi, Japan
| |
Collapse
|
5
|
Voutouri C, Hardin CC, Naranbhai V, Nikmaneshi MR, Khandekar MJ, Gainor JF, Munn LL, Jain RK, Stylianopoulos T. Dynamic heterogeneity in COVID-19: Insights from a mathematical model. PLoS One 2024; 19:e0301780. [PMID: 38820409 PMCID: PMC11142552 DOI: 10.1371/journal.pone.0301780] [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: 11/15/2023] [Accepted: 03/20/2024] [Indexed: 06/02/2024] Open
Abstract
Critical illness, such as severe COVID-19, is heterogenous in presentation and treatment response. However, it remains possible that clinical course may be influenced by dynamic and/or random events such that similar patients subject to similar injuries may yet follow different trajectories. We deployed a mechanistic mathematical model of COVID-19 to determine the range of possible clinical courses after SARS-CoV-2 infection, which may follow from specific changes in viral properties, immune properties, treatment modality and random external factors such as initial viral load. We find that treatment efficacy and baseline patient or viral features are not the sole determinant of outcome. We found patients with enhanced innate or adaptive immune responses can experience poor viral control, resolution of infection or non-infectious inflammatory injury depending on treatment efficacy and initial viral load. Hypoxemia may result from poor viral control or ongoing inflammation despite effective viral control. Adaptive immune responses may be inhibited by very early effective therapy, resulting in viral load rebound after cessation of therapy. Our model suggests individual disease course may be influenced by the interaction between external and patient-intrinsic factors. These data have implications for the reproducibility of clinical trial cohorts and timing of optimal treatment.
Collapse
Affiliation(s)
- Chrysovalantis Voutouri
- Department of Radiation Oncology, Edwin L Steele Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
- Department of Mechanical and Manufacturing Engineering, Cancer Biophysics Laboratory, University of Cyprus, Nicosia, Cyprus
| | - C. Corey Hardin
- Department of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Vivek Naranbhai
- Department of Medicine, Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, United States of America
- Dana-Farber Cancer Institute, Boston, MA, United States of America
- Center for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Mohammad R. Nikmaneshi
- Department of Radiation Oncology, Edwin L Steele Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Melin J. Khandekar
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Justin F. Gainor
- Department of Medicine, Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Lance L. Munn
- Department of Radiation Oncology, Edwin L Steele Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Rakesh K. Jain
- Department of Radiation Oncology, Edwin L Steele Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Triantafyllos Stylianopoulos
- Department of Mechanical and Manufacturing Engineering, Cancer Biophysics Laboratory, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
6
|
Voutouri C, Hardin CC, Naranbhai V, Nikmaneshi MR, Khandekar MJ, Gainor JF, Stylianopoulos T, Munn LL, Jain RK. In silico clinical studies for optimal COVID-19 vaccination schedules in patients with cancer. Cell Rep Med 2024; 5:101436. [PMID: 38508146 PMCID: PMC10982978 DOI: 10.1016/j.xcrm.2024.101436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/25/2023] [Accepted: 01/29/2024] [Indexed: 03/22/2024]
Abstract
This study introduces a tailored COVID-19 model for patients with cancer, incorporating viral variants and immune-response dynamics. The model aims to optimize vaccination strategies, contributing to personalized healthcare for vulnerable groups.
Collapse
Affiliation(s)
- Chrysovalantis Voutouri
- Edwin L Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - C Corey Hardin
- Department of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Vivek Naranbhai
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA; Center for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Mohammad R Nikmaneshi
- Edwin L Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Melin J Khandekar
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Justin F Gainor
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Lance L Munn
- Edwin L Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Samanta S, Banerjee J, Das A, Das S, Ahmed R, Das S, Pal A, Ali KM, Mukhopadhyay R, Giri B, Dash SK. Enhancing Immunological Memory: Unveiling Booster Doses to Bolster Vaccine Efficacy Against Evolving SARS-CoV-2 Mutant Variants. Curr Microbiol 2024; 81:91. [PMID: 38311669 DOI: 10.1007/s00284-023-03597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
A growing number of re-infections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in previously immunized individuals has sparked discussions about the potential need for a booster vaccine dosage to counteract declining antibody levels and new strains. The protective immunity produced by vaccinations, and past illnesses relies on immunological memory. CD4 + T cells, CD8 + T cells, B cells, and long-lasting antibody responses are all components of the adaptive immune system that can generate and maintain this immunological memory. Since novel mutant variants have emerged one after the other, the world has been hit by repeated waves. Various vaccine formulations against SARS-CoV-2 have been administered across the globe. Thus, estimating the efficacy of those vaccines against gradually developed mutant stains is the essential parameter regarding the fate of those vaccine formulations and the necessity of booster doses and their frequency. In this review, focus has also been given to how vaccination stacks up against moderate and severe acute infections in terms of the longevity of the immune cells, neutralizing antibody responses, etc. However, hybrid immunity shows a greater accuracy of re-infection of variants of concern (VOCs) of SARS-CoV-2 than infection and immunization. The review conveys knowledge of detailed information about several marketed vaccines and the status of their efficacy against specific mutant strains of SARS-CoV-2. Furthermore, this review discusses the status of immunological memory after infection, mixed infection, and vaccination.
Collapse
Affiliation(s)
- Sovan Samanta
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Aparna Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sourav Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Rubai Ahmed
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Swarnali Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Amitava Pal
- Department of Physiology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Kazi Monjur Ali
- Department of Nutrition, Maharajadhiraj Uday Chand Women's College, B.C. Road, Bardhaman, 713104, West Bengal, India
| | - Rupanjan Mukhopadhyay
- Department of Physiology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India.
| |
Collapse
|
8
|
Desikan R, Germani M, van der Graaf PH, Magee M. A Quantitative Clinical Pharmacology-Based Framework For Model-Informed Vaccine Development. J Pharm Sci 2024; 113:22-32. [PMID: 37924975 DOI: 10.1016/j.xphs.2023.10.043] [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: 09/06/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/06/2023]
Abstract
Historically, vaccine development and dose optimization have followed mostly empirical approaches without clinical pharmacology and model-informed approaches playing a major role, in contrast to conventional drug development. This is attributed to the complex cascade of immunobiological mechanisms associated with vaccines and a lack of quantitative frameworks for extracting dose-exposure-efficacy-toxicity relationships. However, the Covid-19 pandemic highlighted the lack of sufficient immunogenicity due to suboptimal vaccine dosing regimens and the need for well-designed, model-informed clinical trials which enhance the probability of selection of optimal vaccine dosing regimens. In this perspective, we attempt to develop a quantitative clinical pharmacology-based approach that integrates vaccine dose-efficacy-toxicity across various stages of vaccine development into a unified framework that we term as model-informed vaccine dose-optimization and development (MIVD). We highlight scenarios where the adoption of MIVD approaches may have a strategic advantage compared to conventional practices for vaccines.
Collapse
Affiliation(s)
- Rajat Desikan
- Clinical Pharmacology Modelling & Simulation, GSK, United Kingdom.
| | | | - Piet H van der Graaf
- Certara QSP, Canterbury Innovation Centre, University Road, Canterbury CT2 7FG, United Kingdom; Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Mindy Magee
- Clinical Pharmacology Modelling & Simulation, GSK, United States
| |
Collapse
|
9
|
Piltch-Loeb R, Penrose K, Stanton E, Parcesepe AM, Shen Y, Fleary SA, Nash D. Safety, Efficacy, and Ill Intent: Examining COVID-19 Vaccine Perceptions among the New Undervaccinated Moveable Middle in a U.S. Cohort, October 2022. Vaccines (Basel) 2023; 11:1665. [PMID: 38005997 PMCID: PMC10675675 DOI: 10.3390/vaccines11111665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Individuals who received their primary vaccine series only (with no subsequent booster) may be a new type of "moveable middle" given their receipt of the original COVID-19 vaccination. One population within the moveable middle for whom tailored interventions may be needed is individuals with common mental disorders (CMD). The purpose of this paper is to understand the vaccine perceptions among this new moveable middle-the undervaccinated-and within the undervaccinated to examine the extent to which COVID-19 vaccine perceptions and motivations differ among those with and without symptoms of CMD. Using data from the CHASING COVID Cohort, we examine the relationship between vaccination status, CMD, and vaccine perceptions in the undervaccinated. Among 510 undervaccinated participants who had completed the primary vaccine series but were not boosted, the most common reasons for undervaccination focused on efficacy (not seeing a need for an additional dose, 42.4%; there not being enough evidence that a booster dose is effective, 26.5%; already having had COVID-19, 19.6%). Other concerns were related to safety (long-term side effects, 21.0%; short-term side effects, 17.6%) and logistics (plan to get a booster but haven't had time yet, 18.8%). Overall, the greatest vaccine concerns (over 30%) for the undervaccinated focused on efficacy and safety issues. Symptoms of depression or anxiety were associated with lower levels of vaccine efficacy and greater safety concerns in adjusted models. The implications of our study are that campaigns that are hoping to maximize vaccination uptake should consider focusing on and emphasizing messaging on efficacy and safety issues.
Collapse
Affiliation(s)
- Rachael Piltch-Loeb
- Department of Environmental Occupational and Geospatial Health Sciences, Graduate School of Public Health and Health Policy, City University of New York (CUNY), New York, NY 10027, USA
- Institute for Implementation Science in Population Health (ISPH), City University of New York (CUNY), New York, NY 10027, USA
- Emergency Preparedness Research Evaluation and Practice Program, Harvard T.H. Chan School of Public Health, Boston, MA 02120, USA
| | - Kate Penrose
- Institute for Implementation Science in Population Health (ISPH), City University of New York (CUNY), New York, NY 10027, USA
| | - Eva Stanton
- Emergency Preparedness Research Evaluation and Practice Program, Harvard T.H. Chan School of Public Health, Boston, MA 02120, USA
| | - Angela M. Parcesepe
- Department of Maternal and Child Health, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Yanhan Shen
- Institute for Implementation Science in Population Health (ISPH), City University of New York (CUNY), New York, NY 10027, USA
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, City University of New York (CUNY), New York, NY 10027, USA
| | - Sasha A. Fleary
- Institute for Implementation Science in Population Health (ISPH), City University of New York (CUNY), New York, NY 10027, USA
- Department of Community Health and Social Sciences, Graduate School of Public Health and Health Policy, City University of New York (CUNY), New York, NY 10027, USA
| | - Denis Nash
- Institute for Implementation Science in Population Health (ISPH), City University of New York (CUNY), New York, NY 10027, USA
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, City University of New York (CUNY), New York, NY 10027, USA
| |
Collapse
|
10
|
Dari A, Jacqmin P, Iwaki Y, Neyens M, Le Gars M, Sadoff J, Hardt K, Ruiz‐Guiñazú J, Pérez‐Ruixo JJ. Mechanistic modeling projections of antibody persistence after homologous booster regimens of COVID-19 vaccine Ad26.COV2.S in humans. CPT Pharmacometrics Syst Pharmacol 2023; 12:1485-1498. [PMID: 37715342 PMCID: PMC10583247 DOI: 10.1002/psp4.13025] [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: 06/21/2023] [Accepted: 07/25/2023] [Indexed: 09/17/2023] Open
Abstract
Mechanistic model-based simulations can be deployed to project the persistence of humoral immune response following vaccination. We used this approach to project the antibody persistence through 24 months from the data pooled across five clinical trials in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-seronegative participants following vaccination with Ad26.COV2.S (5 × 1010 viral particles), given either as a single-dose or a homologous booster regimen at an interval of 2, 3, or 6 months. Antibody persistence was quantified as the percentage of participants with detectable anti-spike binding and wild-type virus neutralizing antibodies. The projected overall 24-month persistence after single-dose Ad26.COV2.S was 70.5% for binding antibodies and 55.2% for neutralizing antibodies, and increased after any homologous booster regimen to greater than or equal to 89.9% for binding and greater than or equal to 80.0% for neutralizing antibodies. The estimated model parameters quantifying the rates of antibody production attributed to short-lived and long-lived plasma cells decreased with increasing age, whereas the rate of antibody production mediated by long-lived plasma cells was higher in women relative to men. Accordingly, a more pronounced waning of antibody responses was predicted in men aged greater than or equal to 60 years and was markedly attenuated following any homologous boosting regimen. The findings suggest that homologous boosting might be a viable strategy for maintaining protective effects of Ad26.COV2.S for up to 24 months following prime vaccination. The estimation of mechanistic modeling parameters identified the long-lived plasma cell pathway as a key contributor mediating antibody persistence following single-dose and homologous booster vaccination with Ad26.COV2.S in different subgroups of recipients stratified by age and sex.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research & DevelopmentBeerseBelgium
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Dogra P, Schiavone C, Wang Z, Ruiz-Ramírez J, Caserta S, Staquicini DI, Markosian C, Wang J, Sostman HD, Pasqualini R, Arap W, Cristini V. A modeling-based approach to optimize COVID-19 vaccine dosing schedules for improved protection. JCI Insight 2023; 8:e169860. [PMID: 37227783 PMCID: PMC10371350 DOI: 10.1172/jci.insight.169860] [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: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 05/27/2023] Open
Abstract
While the development of different vaccines slowed the dissemination of SARS-CoV-2, the occurrence of breakthrough infections has continued to fuel the COVID-19 pandemic. To secure at least partial protection in the majority of the population through 1 dose of a COVID-19 vaccine, delayed administration of boosters has been implemented in many countries. However, waning immunity and emergence of new variants of SARS-CoV-2 suggest that such measures may induce breakthrough infections due to intermittent lapses in protection. Optimizing vaccine dosing schedules to ensure prolonged continuity in protection could thus help control the pandemic. We developed a mechanistic model of immune response to vaccines as an in silico tool for dosing schedule optimization. The model was calibrated with clinical data sets of acquired immunity to COVID-19 mRNA vaccines in healthy and immunocompromised participants and showed robust validation by accurately predicting neutralizing antibody kinetics in response to multiple doses of COVID-19 mRNA vaccines. Importantly, by estimating population vulnerability to breakthrough infections, we predicted tailored vaccination dosing schedules to minimize breakthrough infections, especially for immunocompromised individuals. We identified that the optimal vaccination schedules vary from CDC-recommended dosing, suggesting that the model is a valuable tool to optimize vaccine efficacy outcomes during future outbreaks.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Carmine Schiavone
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Zhihui Wang
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz-Ramírez
- Centro de Ciencias de la Salud, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Sergio Caserta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Christopher Markosian
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Surgery, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - H. Dirk Sostman
- Weill Cornell Medicine, New York, New York, USA
- Houston Methodist Research Institute, Houston, Texas, USA
- Houston Methodist Academic Institute, Houston, Texas, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
12
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
13
|
Vicenti I, Basso M, Pirola N, Bragato B, Rossi MC, Giobbia M, Pascoli S, Vinci A, Caputo S, Varasi I, Biba C, Fiaschi L, Zazzi M, Parisi SG. SARS-CoV-2 Neutralizing Antibodies to B.1 and to BA.5 Variant after Booster Dose of BNT162b2 Vaccine in HIV Patients COVID-Naïve and on Successful Antiretroviral Therapy. Vaccines (Basel) 2023; 11:vaccines11040871. [PMID: 37112782 PMCID: PMC10144758 DOI: 10.3390/vaccines11040871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Live virus neutralization is the gold standard to investigate immunity. This prospective observational study aimed to determine the magnitude of response against the original B.1 lineage and against the BA.5 lineage six months after the third BNT162b2 mRNA vaccine dose in patients with HIV infection on successful antiretroviral treatment and no previous SARS-CoV-2 infection. A total of 100 subjects (M/F 83/17, median age 54 years) were included in the analysis: 95 had plasma HIV RNA <40 copies/mL, the median CD4+ T cell count at the administration of the third dose was 580 cells/mm3, and the median nadir CD4+ T cell count was 258 cells/mm3. Neutralizing antibodies (NtAb) against B.1 were detectable in all the subjects, but those to BA.5 were only detected in 88 (p < 0.001). The median NtAb titer to B.1 was significantly higher than that to BA.5 (393 vs. 60, p < 0.0001), and there was a strong positive correlation between the paired measurements (p < 0.0001). Linear regression on a subset of 87 patients excluding outlier NtAb titers showed that 48% of the changes in NtAb titers to BA.5 are related to the changes in value titers to B.1. SARS-CoV-2 variants evolve rapidly, challenging the efficacy of vaccines, and data on comparative NtAb responses may help in tailoring intervals between vaccine doses and in predicting vaccine efficacy.
Collapse
Affiliation(s)
- Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Monica Basso
- Department of Molecular Medicine, University of Padova, 35100 Padova, Italy
| | - Nicole Pirola
- Department of Molecular Medicine, University of Padova, 35100 Padova, Italy
| | - Beatrice Bragato
- Department of Molecular Medicine, University of Padova, 35100 Padova, Italy
| | | | - Mario Giobbia
- Infectious Diseases Unit, Treviso Hospital, 31100 Treviso, Italy
| | - Susanna Pascoli
- Microbiology Unit, Department of Specialist and Laboratory Medicine, Ca' Foncello University Hospital, 31100 Treviso, Italy
| | - Antonio Vinci
- Hospital Health Management Area, Local Health Authority "Roma 1", Borgo Santo Spirito 3, 00193 Rome, Italy
| | - Sara Caputo
- Department of Molecular Medicine, University of Padova, 35100 Padova, Italy
| | - Ilenia Varasi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Camilla Biba
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Lia Fiaschi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
14
|
Lasagna A, Cassaniti I, Arena F, Bergami F, Percivalle E, Comolli G, Sarasini A, Ferrari A, Cicognini D, Schiavo R, Lo Cascio G, Pedrazzoli P, Baldanti F. Persistence of Immune Response Elicited by Three Doses of mRNA Vaccine against SARS-CoV-2 in a Cohort of Patients with Solid Tumors: A One-Year Follow-Up. Int J Mol Sci 2023; 24:ijms24076731. [PMID: 37047704 PMCID: PMC10095115 DOI: 10.3390/ijms24076731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
The role and durability of the immunogenicity of the BNT162b2 mRNA vaccine against severe acute respiratory virus 2 (SARS-CoV-2), in cancer patients one year after receiving the third dose have to be elucidated. We have prospectively evaluated the long-term immunogenicity of the third dose of the SARS-CoV-2 BNT162b2 mRNA vaccine in 55 patients undergoing active treatment. Neutralizing antibody (NT Ab) titers against Omicron variants and total anti-trimeric S IgG levels were measured one year after the third dose. Heparinized whole-blood samples were used for the assessment of the SARS-CoV-2 interferon-γ release assay (IGRA). Thirty-seven patients (67.3%) showed positive total anti-trimeric S IgG one year after the third dose. Looking at the T-cell response against the spike protein, the frequency of responder patients did not decrease significantly between six and twelve months after the third dose. Finally, less than 20% of cancer patients showed an undetectable NT Ab titer against BA.1 and BA.5 variants of concern (VOCs). Underlying therapies seem to not affect the magnitude or frequency of the immune response. Our work underlines the persistence of humoral and cellular immune responses against BNT162b2 in a cohort of cancer patients one year after receiving the third dose, regardless of the type of underlying therapy.
Collapse
Affiliation(s)
- Angioletta Lasagna
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Irene Cassaniti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Francesca Arena
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Federica Bergami
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Elena Percivalle
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giuditta Comolli
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Antonella Sarasini
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Alessandro Ferrari
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Daniela Cicognini
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Roberta Schiavo
- Microbiology Unit, Hospital Guglielmo da Saliceto, 29121 Piacenza, Italy
| | - Giuliana Lo Cascio
- Microbiology Unit, Hospital Guglielmo da Saliceto, 29121 Piacenza, Italy
| | - Paolo Pedrazzoli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Internal Medicine and Medical Therapy, University of Pavia, 27100 Pavia, Italy
| | - Fausto Baldanti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
15
|
Pedrazzoli P, Lasagna A, Cassaniti I, Piralla A, Squeri A, Bruno R, Sacchi P, Baldanti F, Di Maio M, Beretta GD, Cinieri S, Silvestris N. Vaccination for seasonal flu, pneumococcal infection, and SARS-CoV-2 in patients with solid tumors: recommendations of the Associazione Italiana di Oncologia Medica (AIOM). ESMO Open 2023; 8:101215. [PMID: 37104930 PMCID: PMC10067463 DOI: 10.1016/j.esmoop.2023.101215] [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: 03/03/2023] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Patients with cancer have a well-known and higher risk of vaccine-preventable diseases (VPDs). VPDs may cause severe complications in this setting due to the immune system impairment, malnutrition and oncological treatments. Despite this evidence, vaccination rates are inadequate. The Italian Association of Medical Oncology (AIOM) has been involved in vaccination awareness since 2014. Based on a careful review of the available data about the immunogenicity, effectiveness and safety of flu, pneumococcal and anti-SARS-CoV-2 vaccines, we report the recommendations of the Associazione Italiana di Oncologia Medica about these vaccinations in adult patients with solid tumors. AIOM recommends comprehensive education on the issue of VPDs. We believe that a multidisciplinary care model may improve the vaccination coverage in immunocompromised patients. Continued surveillance, implementation of preventive practices and future well-designed immunological prospective studies are essential for a better management of our patients with cancer.
Collapse
Affiliation(s)
- P Pedrazzoli
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy; Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Lasagna
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - I Cassaniti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Piralla
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Squeri
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy; School of Specialization in Medical Oncology, University of Messina, Messina, Italy
| | - R Bruno
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Division of Infectious Diseases I, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - P Sacchi
- Division of Infectious Diseases I, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - F Baldanti
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - M Di Maio
- Department of Oncology, University of Turin, Division of Medical Oncology, Ordine Mauriziano Hospital, Turin, Italy
| | - G D Beretta
- Medical Oncology Unit, Santo Spirito Hospital, Pescara, Italy
| | - S Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | - N Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| |
Collapse
|