1
|
Barrington H, McCabe TJD, Donnachie K, Fyfe C, McFall A, Gladkikh M, McGuire J, Yan C, Reid M. Parallel and High Throughput Reaction Monitoring with Computer Vision. Angew Chem Int Ed Engl 2025; 64:e202413395. [PMID: 39166494 DOI: 10.1002/anie.202413395] [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: 07/16/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
We report the development and applications of a computer vision based reaction monitoring method for parallel and high throughput experimentation (HTE). Whereas previous efforts reported methods to extract bulk kinetics of one reaction from one video, this new approach enables one video to capture bulk kinetics of multiple reactions running in parallel. Case studies, in and beyond well-plate high throughput settings, are described. Analysis of parallel dye-quenching hydroxylations, DMAP-catalysed esterification, solid-liquid sedimentation dynamics, metal catalyst degradation, and biologically-relevant sugar-mediated nitro reduction reactions have each provided insight into the scope and limitations of camera-enabled high throughput kinetics as a means of widening known analytical bottlenecks in HTE for reaction discovery, mechanistic understanding, and optimisation. It is envisaged that the nature of the multi-reaction time-resolved datasets made available by this analytical approach will later serve a broad range of downstream efforts in machine learning approaches to exploring chemical space.
Collapse
Affiliation(s)
- H Barrington
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - T J D McCabe
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - K Donnachie
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Calum Fyfe
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - A McFall
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - M Gladkikh
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - J McGuire
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - C Yan
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - M Reid
- Department of Pure & Applied Chemistry, University of Strathclyde, Glasgow, UK
| |
Collapse
|
2
|
Piliper EA, Reed JC, Greninger AL. Clinical validation of an RSV neutralization assay and analysis of cross-sectional sera associated with 2021-2023 RSV outbreaks to investigate the immunity debt hypothesis. Microbiol Spectr 2024; 12:e0211524. [PMID: 39470275 PMCID: PMC11619414 DOI: 10.1128/spectrum.02115-24] [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: 08/23/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of acute respiratory infections and hospitalization in infants and the elderly. Newly approved vaccines and the prophylactic antibody nirsevimab have heightened interest in RSV immunologic surveillance, necessitating the development of high-throughput assays assessing anti-RSV neutralizing activity. Quantitative viral neutralization remains the best correlate of protection for RSV infection and the gold standard for RSV immunological testing. Here, we developed a high-throughput RSV strain A2 focus-reduction neutralization test validated to Clinical Laboratory Improvement Amendments (CLIA)/ Good Clinical Laboratory Practices (GCLP) standards using both clinical specimens and commercially available reference sera. The assay is highly accurate, generating reference serum neutralizing titers within twofold of established assays, with an analytical measurement range between 8 and 1,798 international units per mL (IU/mL). Neutralizing activity measured by the assay strongly correlated with antibody titer determined via indirect enzyme-linked immunosorbent assay (ELISA) (ρ = 1.0, P = 0.0014). Individuals recently having tested positive via quantitative reverse transcription polymerase chain reaction (RT-qPCR) for RSV had a 9.1-fold higher geometric mean neutralizing titer relative to RSV PCR negatives (P-value = 0.09). The validated assay was then used to investigate the immunity debt hypothesis for resurgent RSV outbreaks in the 2022-2023 season, using adult clinical remnant sera sent for herpes simplex virus (HSV)-1/2 antibody testing. There was no difference in geometric mean anti-RSV neutralizing titers between sera sampled before and after the 2022-2023 RSV outbreak (P = 0.68). These data are consistent with limited changes in RSV-neutralizing antibody levels in adults across the 2022-23 RSV outbreak. IMPORTANCE Population surveillance studies of serum-neutralizing activity against RSV are crucial for evaluating RSV vaccine efficacy and vulnerabilities to new strains. Here, we designed and validated a high-throughput assay for assessing anti-RSV neutralizing activity, standardized its measurements for comparison with other methodologies, and demonstrated its applicability to real-world samples. Our assay is precise, linear, and yields measurements consistent with other standardized assays, offering a methodology useful for large-scale studies of RSV immunity. We also find no significant difference in neutralizing titers among adults between those taken before and after large RSV outbreaks associated with the latter stages of the coronavirus disease of 2019 (COVID-19) public health emergency, underlining the need for a greater understanding of the dynamics of serological responses to RSV infection.
Collapse
Affiliation(s)
- Eli A. Piliper
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
| | - Jonathan C. Reed
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
| | - Alexander L. Greninger
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
3
|
Nham E, Kim J, Lee J, Park H, Kim J, Lee S, Choi J, Kim KT, Yoon JG, Hwang SY, Song JY, Cheong HJ, Kim WJ, Park MS, Noh JY. Low Neutralizing Activities to the Omicron Subvariants BN.1 and XBB.1.5 of Sera From the Individuals Vaccinated With a BA.4/5-Containing Bivalent mRNA Vaccine. Immune Netw 2023; 23:e43. [PMID: 38188597 PMCID: PMC10767551 DOI: 10.4110/in.2023.23.e43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/09/2023] [Accepted: 10/30/2023] [Indexed: 01/09/2024] Open
Abstract
The continuous emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants has provided insights for updating current coronavirus disease 2019 (COVID-19) vaccines. We examined the neutralizing activity of Abs induced by a BA.4/5-containing bivalent mRNA vaccine against Omicron subvariants BN.1 and XBB.1.5. We recruited 40 individuals who had received a monovalent COVID-19 booster dose after a primary series of COVID-19 vaccinations and will be vaccinated with a BA.4/5-containing bivalent vaccine. Sera were collected before vaccination, one month after, and three months after a bivalent booster. Neutralizing Ab (nAb) titers were measured against ancestral SARS-CoV-2 and Omicron subvariants BA.5, BN.1, and XBB.1.5. BA.4/5-containing bivalent vaccination significantly boosted nAb levels against both ancestral SARS-CoV-2 and Omicron subvariants. Participants with a history of SARS-CoV-2 infection had higher nAb titers against all examined strains than the infection-naïve group. NAb titers against BN.1 and XBB.1.5 were lower than those against the ancestral SARS-CoV-2 and BA.5 strains. These results suggest that COVID-19 vaccinations specifically targeting emerging Omicron subvariants, such as XBB.1.5, may be required to ensure better protection against SARS-CoV-2 infection, especially in high-risk groups.
Collapse
Affiliation(s)
- Eliel Nham
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Jineui Kim
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Jungmin Lee
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Heedo Park
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Jeonghun Kim
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Sohyun Lee
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Jaeuk Choi
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Kyung Taek Kim
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Jin Gu Yoon
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Soon Young Hwang
- Department of Biostatistics, Korea University College of Medicine, Seoul 02841, Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Woo Joo Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Man-Seong Park
- Department of Microbiology, Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| |
Collapse
|
4
|
Nussbaum J, Cao X, Railkar RA, Sachs JR, Spellman DS, Luk J, Shaw CA, Cejas PJ, Citron MP, Al-Ibrahim M, Han D, Pagnussat S, Stoch SA, Lai E, Bett AJ, Espeseth AS. Evaluation of a stabilized RSV pre-fusion F mRNA vaccine: Preclinical studies and Phase 1 clinical testing in healthy adults. Vaccine 2023; 41:6488-6501. [PMID: 37777449 DOI: 10.1016/j.vaccine.2023.05.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/15/2023] [Accepted: 05/26/2023] [Indexed: 10/02/2023]
Abstract
Human respiratory syncytial virus (RSV) causes a substantial proportion of respiratory tract infections worldwide. Although RSV reinfections occur throughout life, older adults, particularly those with underlying comorbidities, are at risk for severe complications from RSV. There is no RSV vaccine available to date, and treatment of RSV in adults is largely supportive. A correlate of protection for RSV has not yet been established, but antibodies targeting the pre-fusion conformation of the RSV F glycoprotein play an important role in RSV neutralization. We previously reported a Phase 1 study of an mRNA-based vaccine (V171) expressing a pre-fusion-stabilized RSV F protein (mDS-Cav1) in healthy adults. Here, we evaluated an mRNA-based vaccine (V172) expressing a further stabilized RSV pre-fusion F protein (mVRC1). mVRC1 is a single chain version of RSV F with interprotomer disulfides in addition to the stabilizing mutations present in the mDS-Cav1 antigen. The immunogenicity of the two mRNA-based vaccines encoding mVRC1 (V172) or a sequence-optimized version of mDS-Cav1 to improve transcriptional fidelity (V171.2) were compared in RSV-naïve and RSV-experienced African green monkeys (AGMs). V172 induced higher neutralizing antibody titers than V171.2 and demonstrated protection in the AGM challenge model. We conducted a Phase 1, randomized, placebo-controlled, clinical trial of 25 μg, 100 μg, 200 μg, or 300 μg of V172 in healthy older adults (60-79 years old; N = 112) and 100 μg, 200 μg, or 300 μg of V172 in healthy younger adults (18-49 years old; N = 48). The primary clinical objectives were to evaluate the safety and tolerability of V172, and the secondary objective was to evaluate RSV serum neutralization titers. The most commonly reported solicited adverse events were injection-site pain, injection-site swelling, headache, and tiredness. V172 was generally well tolerated in older and younger adults and increased serum neutralizing antibody titers, pre-fusion F-specific competing antibody titers, and RSV F-specific T-cell responses.
Collapse
Affiliation(s)
| | - Xin Cao
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Virus reduction neutralization test and LI-COR microneutralization assay bridging and WHO international standard calibration studies for respiratory syncytial virus. Bioanalysis 2022; 14:1067-1079. [PMID: 36125040 DOI: 10.4155/bio-2022-0097] [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: 11/17/2022] Open
Abstract
Background: Respiratory syncytial virus (RSV) vaccine is an unmet medical need. The virus reduction neutralization test (VRNT) was developed to replace the LI-COR microneutralization assay to measure RSV neutralization titers. Methods: A bridging study using selected V171 phase I samples and calibration studies using the WHO international standard antiserum to RSV were performed to compare VRNT and LI-COR. Results: From the bridging study, we showed good concordance between VRNT and LI-COR titers, and similar post-/pre-vaccination titer ratios. From the calibration studies, we can convert VRNT and LI-COR titers into similar IU/ml. Conclusion: The VRNT and LI-COR microneutralization assay correlate well and the titers can be standardized as similar IU/ml, enabling direct comparison of titers from different assays.
Collapse
|
6
|
Orito Y, Otani N, Matsumoto Y, Fujimoto K, Oshima N, Maas BM, Caro L, Aliprantis AO, Cox KS, Tokumaru O, Kodama M, Kudo H, Imai H, Uemura N. A Phase 1 Study to Evaluate Safety, Pharmacokinetics, and Pharmacodynamics of Respiratory Syncytial Virus (RSV) Neutralizing Monoclonal Antibody MK-1654 in Healthy Japanese Adults. Clin Transl Sci 2022; 15:1753-1763. [PMID: 35506164 PMCID: PMC9283748 DOI: 10.1111/cts.13290] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/31/2022] [Accepted: 04/14/2022] [Indexed: 12/05/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among all infants worldwide and remains a significant cause of morbidity and mortality. To address this unmet medical need, MK‐1654, a half‐life extended RSV neutralizing monoclonal antibody, is in clinical development for the prevention of RSV disease in infants. This was a phase I, randomized, placebo‐controlled, single‐site, double‐blind trial of MK‐1654 in 44 healthy Japanese adults. The safety, tolerability, pharmacokinetics, antidrug antibodies (ADAs), and serum neutralizing antibody (SNA) titers against RSV were evaluated for 1 year after a single intramuscular (i.m.) or intravenous (i.v.) dose of MK‐1654 or placebo in five groups (100 mg i.m., 300 mg i.m., 300 mg i.v., 1000 mg i.v., or placebo). MK‐1654 was generally well‐tolerated in Japanese adults. There were no serious drug‐related adverse events (AEs) reported in any MK‐1654 recipient and no discontinuations due to any AEs in the study. The half‐life of MK‐1654 ranged from 76 to 91 days across dosing groups. Estimated bioavailability was 86% for 100 mg i.m. and 77% for 300 mg i.m. One participant out of 33 (3.0%) developed detectable ADA with no apparent associated AEs. The RSV SNA titers increased in a dose‐dependent manner among participants who received MK‐1654. These data support the development of MK‐1654 for use in Japanese infants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Antonios O Aliprantis
- Merck and Co., Inc., Kenilworth, NJ, USA.,Present address: Flagship Pioneering, Boston, MA, USA
| | - Kara S Cox
- Merck and Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | | |
Collapse
|
7
|
Sun D, Hsu A, Quiroz J, He X, Whiteman MC, Gurney KB, Dellatore S. Development and comparison of three cell-based potency assays for anti-respiratory syncytial virus monoclonal antibody. Biologicals 2021; 74:1-9. [PMID: 34716091 DOI: 10.1016/j.biologicals.2021.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/21/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022] Open
Abstract
There is an increasing demand for monoclonal antibody (mAb) therapies to confer passive immunity against viral diseases. Respiratory syncytial virus (RSV) is the most common cause of bronchiolitis, lower respiratory tract infections, and hospitalization in infants. Currently, there is no RSV vaccine but a humanized mAb available for high risk infants. MK-1654 is a fully human mAb with YTE mutation in the fragment crystallizable (Fc) region to extend the half-life in circulation. It binds to a highly conserved epitope of RSV Fusion protein with high affinity and neutralizes RSV infection. A functional cell-based assay is a regulatory requirement for clinical development, commercial release, and stability testing of MK-1654. In this study, we have evaluated three RSV neutralization assays to test the potency of MK-1654, including an imaging-based virus reduction neutralization test (VRNT) and two reporter virus-based assays (RSV-GFP and RSV-NLucP). All three methods showed good dose response curves of MK-1654 with similar EC50 values. RSV-NLucP method was chosen for further development because it is simple and can be easily adapted to quality control testing laboratories. After optimization, the RSV-NLucP assay was pre-qualified with good linearity, relative accuracy, intermediate precision, and specificity, therefore suitable for a cell-based potency assay.
Collapse
Affiliation(s)
- Dengyun Sun
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA.
| | - Amy Hsu
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jorge Quiroz
- Research Chemistry Manufacturing & Controls Statistics, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Xi He
- Infectious Disease and Vaccines, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Melissa C Whiteman
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Kevin B Gurney
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Shara Dellatore
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| |
Collapse
|