158 results on '"Annaliesa S. Anderson"'
Search Results
2. Bivalent Prefusion F Vaccine in Pregnancy to Prevent RSV Illness in Infants
- Author
-
Beate Kampmann, Shabir A. Madhi, Iona Munjal, Eric A.F. Simões, Barbara A. Pahud, Conrado Llapur, Jeffrey Baker, Gonzalo Pérez Marc, David Radley, Emma Shittu, Julia Glanternik, Hasra Snaggs, James Baber, Philip Zachariah, Shaun L. Barnabas, Merlin Fausett, Tyler Adam, Nicole Perreras, Marlies A. Van Houten, Anu Kantele, Li-Min Huang, Louis J. Bont, Takeo Otsuki, Sergio L. Vargas, Joanna Gullam, Bruce Tapiero, Renato T. Stein, Fernando P. Polack, Heather J. Zar, Nina B. Staerke, María Duron Padilla, Peter C. Richmond, Kenneth Koury, Katherine Schneider, Elena V. Kalinina, David Cooper, Kathrin U. Jansen, Annaliesa S. Anderson, Kena A. Swanson, William C. Gruber, and Alejandra Gurtman
- Subjects
General Medicine - Published
- 2023
3. Efficacy and Safety of a Bivalent RSV Prefusion F Vaccine in Older Adults
- Author
-
Edward E. Walsh, Gonzalo Pérez Marc, Agnieszka M. Zareba, Ann R. Falsey, Qin Jiang, Michael Patton, Fernando P. Polack, Conrado Llapur, Pablo A. Doreski, Kumar Ilangovan, Mika Rämet, Yasushi Fukushima, Nazreen Hussen, Louis J. Bont, Jose Cardona, Elliot DeHaan, Giselle Castillo Villa, Marinela Ingilizova, Daniel Eiras, Tarek Mikati, Rupal N. Shah, Katherine Schneider, David Cooper, Kenneth Koury, Maria-Maddalena Lino, Annaliesa S. Anderson, Kathrin U. Jansen, Kena A. Swanson, Alejandra Gurtman, William C. Gruber, and Beate Schmoele-Thoma
- Subjects
General Medicine - Published
- 2023
4. Identification of a Novel Keto Sugar Component inStreptococcus pneumoniaeSerotype 12F Capsular Polysaccharide and Impact on Vaccine Immunogenicity
- Author
-
Kelly Sackett, Paul Brown, Kaushik Dutta, Ingrid L. Scully, Seema Gangolli, Kelvin Looi, Sandeep Nemani, Angela Yeou Hsiung Yu, Mark Kleven, Jin Xie, Justin Moran, Michael W. Pride, Annaliesa S. Anderson, and Jason Lotvin
- Subjects
Immunology ,Immunology and Allergy ,Immunotherapy and Vaccines - Abstract
Implementation of conjugate vaccine technology revolutionized the ability to effectively elicit long-lasting immune responses to bacterial capsular polysaccharides. Although expansion of conjugate vaccine serotype coverage is designed to target residual disease burden to pneumococcal serotypes not contained in earlier vaccine versions, details of polysaccharide Ag structure, heterogeneity, and epitope structure components contributing to vaccine-mediated immunity are not always clear. Analysis of Streptococcus pneumoniae serotype 12F polysaccharide by two-dimensional nuclear magnetic resonance spectroscopy and mass spectrometry revealed a partial substitution of N-acetyl-galactosamine by the keto sugar 2-acetamido-2,6-dideoxy-xylo-hexos-4-ulose (Sug) in up to 25% of the repeat units. This substitution was not described in previous published structures for 12F. Screening a series of contemporary 12F strains isolated from humans (n = 17) identified Sug incorporation at varying levels in all strains examined. Thus, partial Sug substitution in S. pneumoniae serotype 12F may have always been present but is now detectable by state-of-the-art analytical techniques. During the steps of conjugation, the serotype 12F Sug epitope is modified by reduction, and both polysaccharide PPSV23 and conjugate PCV20 vaccines contain 12F Ags with little to no Sug epitope. Both PCV20 and PPSV23 vaccines were evaluated for protection against circulating 12F strains with varying amounts of Sug in their repeat unit based on an opsonophagocytic killing assay involving HL-60 cells and rabbit complement. Both vaccines elicited human-derived neutralizing Abs against serotype 12F, independent of Sug level between ∼2 and 25 mol%. These findings suggest that the newly identified serotype 12F Sug epitope is likely not an essential epitope for vaccine-elicited protection.
- Published
- 2023
5. Neutralization of BA.4–BA.5, BA.4.6, BA.2.75.2, BQ.1.1, and XBB.1 with Bivalent Vaccine
- Author
-
Jing Zou, Chaitanya Kurhade, Sohil Patel, Nicholas Kitchin, Kristin Tompkins, Mark Cutler, David Cooper, Qi Yang, Hui Cai, Alexander Muik, Ying Zhang, Dung-Yang Lee, Ugur Şahin, Annaliesa S. Anderson, William C. Gruber, Xuping Xie, Kena A. Swanson, and Pei-Yong Shi
- Subjects
General Medicine - Published
- 2023
6. Safety and immunogenicity of a primary series and booster dose of the meningococcal serogroup B-factor H binding protein vaccine (MenB-FHbp) in healthy children aged 1–9 years: two phase 2 randomised, controlled, observer-blinded studies
- Author
-
Helen S Marshall, Timo Vesikari, Peter C Richmond, Jacek Wysocki, Leszek Szenborn, Johannes Beeslaar, Jason D Maguire, Paul Balmer, Robert O'Neill, Annaliesa S Anderson, Jean-Louis Prégaldien, Roger Maansson, Han-Qing Jiang, and John L Perez
- Subjects
Infectious Diseases - Abstract
The meningococcal serogroup B-factor H binding protein vaccine (MenB-FHbp) is licensed for use in children aged 10 years or older for protection against invasive serogroup B meningococcal disease. Because young children are at increased risk of invasive meningococcal disease, MenB-FHbp clinical data in this population are needed.We conducted two phase 2 randomised, controlled, observer-blinded studies including healthy toddlers (age 12-23 months) across 26 Australian, Czech, Finnish, and Polish centres, and older children (age 2-9 years) across 14 Finnish and Polish centres. Exclusion criteria included previous vaccinations against serogroup B meningococcus or hepatitis A virus (HAV), and chronic antibiotic use. Toddlers were randomly allocated (2:1) via an interactive response technology system to receive either 60 μg or 120 μg MenB-FHbp or HAV vaccine and saline (control). Older children were randomly allocated (3:1) to receive 120 μg MenB-FHbp or control, with stratification by age group (2-3 years and 4-9 years). All vaccinations were administered as three doses (0, 2, and 6 months, with only saline given at 2 months in the control group). Toddlers who received 120 μg MenB-FHbp could receive a 120 μg booster dose 24 months after the end of the primary series. The percentages of participants with serum bactericidal activity using human complement (hSBA) titres at or above the lower limit of quantification (LLOQ; all greater than the 1:4 correlate of protection) against four test strains of serogroup B meningococcus 1 month after the third dose (primary immunogenicity endpoint) were measured in the evaluable immunogenicity populations (participants who received the vaccine as randomised, had available and determinate hSBA results, and had no major protocol violations). Not all participants were tested against all strains because of serum sample volume constraints. The frequencies of reactogenicity and adverse events after each dose were recorded in the safety population (all participants who received at least one dose and had safety data available). These studies are registered with ClinicalTrials.gov (NCT02534935 and NCT02531698) and are completed.Between Aug 31, 2015, and Aug 22, 2016, for the toddler study and between Aug 27, 2015, and March 7, 2016, for the older children study, we enrolled and randomly allocated 396 toddlers (60 μg MenB-FHbp group n=44; 120 μg MenB-FHbp group n=220; control group n=132) and 400 older children (120 μg MenB-FHbp group n=294; control group n=106). 1 month after the third dose, the proportions of participants with hSBA titres at or above the LLOQ ranged across test strains from 85·0% (95% CI 62·1-96·8; 17 of 20 participants) to 100·0% (82·4-100·0; 19 of 19) in toddlers receiving 60 μg MenB-FHbp, and from 71·6% (61·4-80·4; 68 of 95) to 100·0% (96·2-100·0; 95 of 95) in toddlers receiving 120 μg MenB-FHbp, and from 79·1% (71·2-85·6; 106 of 134) to 100·0% (97·4-100·0; 139 of 139) in children aged 2-9 years receiving 120 μg MenB-FHbp. hSBA titres peaked at 1 month after the third primary dose of MenB-FHbp and then declined over time. 24 months after the third dose in the toddler study, the proportions with hSBA titres at or above the LLOQ ranged from 0·0% (0·0-17·6; 0 of 19 participants) to 41·2% (18·4-67·1; seven of 17) in those who received 60 μg MenB-FHbp and from 3·7% (0·8-10·4; three of 81) to 22·8% (14·1-33·6; 18 of 79) in those who received 120 μg MenB-FHbp. 1 month after the booster dose in toddlers, the proportions with hSBA titres at or above the LLOQ were higher than at 1 month after the primary series. MenB-FHbp reactogenicity was mostly transient and of mild to moderate severity. Adverse event frequency was similar between the MenB-FHbp and control groups and less frequent following MenB-FHbp booster than following primary doses. Two participants from the toddler study (both from the 120 μg MenB-FHbp group) and four from the older children study (three from the 120 μg MenB-FHbp group and one from the control group) were withdrawn from the study because of adverse events.MenB-FHbp was well tolerated and induced protective immune responses in a high proportion of participants. These findings support a favourable MenB-FHbp immunogenicity and reactogenicity profile in young children, a population at increased risk of adverse invasive meningococcal disease outcomes.Pfizer.
- Published
- 2023
7. Semisynthetic Glycoconjugate Vaccine Candidates against Escherichia coli O25B Induce Functional IgG Antibodies in Mice
- Author
-
Arun Naini, Max Peter Bartetzko, Someswara Rao Sanapala, Felix Broecker, Victoria Wirtz, Marilda P. Lisboa, Sharavathi G. Parameswarappa, Daniel Knopp, Jessica Przygodda, Matthias Hakelberg, Rosalind Pan, Axay Patel, Laurent Chorro, Arthur Illenberger, Christopher Ponce, Srinivas Kodali, Jacqueline Lypowy, Annaliesa S. Anderson, Robert G. K. Donald, Arne von Bonin, and Claney L. Pereira
- Published
- 2022
8. Streptococcus pneumoniae serotype 15B polysaccharide conjugate elicits a cross-functional immune response against serotype 15C but not 15A
- Author
-
Li Hao, Michelle M. Kuttel, Neil Ravenscroft, Allison Thompson, A. Krishna Prasad, Seema Gangolli, Charles Tan, David Cooper, Wendy Watson, Paul Liberator, Michael W. Pride, Kathrin U. Jansen, Annaliesa S. Anderson, and Ingrid L. Scully
- Subjects
Vaccines, Conjugate ,General Veterinary ,General Immunology and Microbiology ,Immunity ,Public Health, Environmental and Occupational Health ,Serogroup ,Antibodies, Bacterial ,Pneumococcal Infections ,Pneumococcal Vaccines ,Streptococcus pneumoniae ,Infectious Diseases ,Polysaccharides ,Humans ,Molecular Medicine ,Aged - Abstract
Protection conferred by pneumococcal polysaccharide conjugate vaccines (PCVs) is associated with PCV-induced antibodies against vaccine-covered serotypes that exhibit functional opsonophagocytic activity (OPA). Structural similarity between capsular polysaccharides of closely related serotypes may result in induction of cross-reactive antibodies with or without a cross-functional activity against a serotype not covered by a PCV, with the former providing an additional protective clinical benefit. Serotypes 15B, 15A, and 15C, in the serogroup 15, are among the most prevalent Streptococcus pneumoniae serotypes associated with invasive pneumococcal disease following the implementation of a 13-valent PCV; in addition, 15B contributes significantly to acute otitis media. Serological discrimination between closely related serotypes such as 15B and 15C is complicated; here, we implemented an algorithm to quickly differentiate 15B from its closely related serotypes 15C and 15A directly from whole-genome sequencing data. In addition, molecular dynamics simulations of serotypes 15A, 15B, and 15C polysaccharides demonstrated that while 15B and 15C polysaccharides assume rigid branched conformation, 15A polysaccharide assumes a flexible linear conformation. A serotype 15B conjugate, included in a 20-valent PCV (PCV20), induced cross-functional OPA serum antibody responses against the structurally similar serotype 15C but not against serotype 15A, both not included in PCV20. In PCV20-vaccinated adults (18-49 years), robust OPA antibody titers were detected against both serotypes 15B (the geometric mean titer [GMT] of 19,334) and 15C (GMTs of 1692 and 2747 for strains PFE344340 and PFE1160, respectively), but were negligible against serotype 15A (GMTs of 10 and 30 for strains PFE593551 and PFE647449, respectively). Cross-functional 15B/C responses were also confirmed using sera from a larger group of older adults (60-64 years).
- Published
- 2022
9. Calibration of a serum reference standard for Group B streptococcal polysaccharide conjugate vaccine development using surface plasmon resonance
- Author
-
Alexandre Esadze, Christopher D. Grube, Sabine Wellnitz, Suddham Singh, Ha H. Nguyen, Michelle A. Gaylord, Aiping Zhu, Alexey Gribenko, Charles Y. Tan, Annaliesa S. Anderson, and Raphael Simon
- Subjects
Pharmacology ,Infectious Diseases ,Immunology ,Pharmacology (medical) - Abstract
Group B streptococcus (GBS) is a leading cause of neonatal morbidity and mortality worldwide. Development of a maternal vaccine to protect newborns through placentally transferred antibody is considered feasible based on the well-established relationship between anti-GBS capsular polysaccharide (CPS) IgG levels at birth and reduced risk of neonatal invasive GBS. An accurately calibrated serum reference standard that can be used to measure anti-CPS concentrations is critical for estimation of protective antibody levels across serotypes and potential vaccine performance. For this, precise weight-based measurement of anti-CPS IgG in sera is required. Here, we report an improved approach for determining serum anti-CPS IgG levels using surface plasmon resonance with monoclonal antibody standards, coupled with a direct Luminex-based immunoassay. This technique was used to quantify serotype-specific anti-CPS IgG levels in a human serum reference pool derived from subjects immunized with an investigational six-valent GBS glycoconjugate vaccine.
- Published
- 2023
10. Nirmatrelvir–Ritonavir and Viral Load Rebound in Covid-19
- Author
-
Annaliesa S, Anderson, Patrick, Caubel, and James M, Rusnak
- Subjects
Drug Combinations ,Ritonavir ,Lactams ,Proline ,Leucine ,Recurrence ,Nitriles ,Humans ,General Medicine ,Viral Load ,Antiviral Agents ,COVID-19 Drug Treatment - Published
- 2022
11. An oral SARS-CoV-2 M pro inhibitor clinical candidate for the treatment of COVID-19
- Author
-
Nandini Chaturbhai Patel, Dafydd R. Owen, Rhonda D. Cardin, Karen J. Coffman, Jean G. Sathish, Devendra K. Rai, Heather Eng, Charlotte Moira Norfor Allerton, Jack C. Lee, Britton Boras, Melissa Avery, Qingyi Yang, Eugene P. Kadar, Anthony Carlo, Annaliesa S. Anderson, Matthew R. Reese, Li Di, Jisun Lee, Lisa Aschenbrenner, Stephen Noell, Lawrence W. Updyke, Martin Pettersson, Scott A. Gibson, Matthew F. Sammons, Al E. Stewart, Yuao Zhu, Alyssa Dantonio, Stephen W. Mason, Brett L. Hurst, Ketan S. Gajiwala, Claire M. Steppan, Liuqing Wei, Patrick Robert Verhoest, Samantha Elizabeth Greasley, Simon Berritt, R. Scott Obach, RoseAnn Ferre, Ravi Shankar P. Singh, Amit S. Kalgutkar, Jonathan J. Novak, Kevin Ogilvie, Wei Liu, and Jamison B. Tuttle
- Subjects
2019-20 coronavirus outbreak ,Multidisciplinary ,Coronavirus disease 2019 (COVID-19) ,business.industry ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pandemic ,Outbreak ,Medicine ,business ,Virology - Abstract
Path to another drug against COVID-19 The rapid development of vaccines has been crucial in battling the ongoing COVID-19 pandemic. However, access challenges remain, breakthrough infections occur, and emerging variants present increased risk. Developing antiviral therapeutics is therefore a high priority for the treatment of COVID-19. Some drug candidates in clinical trials act against the viral RNA-dependent RNA polymerase, but there are other viral enzymes that have been considered good targets for inhibition by drugs. Owen et al . report the discovery and characterization of a drug against the main protease involved in the cleavage of polyproteins involved in viral replication. The drug, PF-07321332, can be administered orally, has good selectivity and safety profiles, and protects against infection in a mouse model. In a phase 1 clinical trial, the drug reached concentrations expected to inhibit the virus based on in vitro studies. It also inhibited other coronaviruses, including severe acute respiratory syndrome coronavirus 1 and Middle East respiratory syndrome coronavirus, and could be in the armory against future viral threats. —VV
- Published
- 2021
12. LB748. Efficacy And Safety Of Bivalent Respiratory Syncytial Virus (RSVpreF) Vaccine In Older Adults
- Author
-
Edward E Walsh, Fernando Polack, Agnieszka Zareba, Ann R Falsey, Gonzalo Perez Marc, Qin Jiang, Kathy Schneider, David Cooper, Maria Maddalena Lino, Annaliesa S Anderson, Katherin U Jensen, Kena A Swanson, Alejandra C Gurtman, William C Gruber, and Beate Schmoele-Thoma
- Subjects
Infectious Diseases ,Oncology - Abstract
Background Respiratory syncytial virus (RSV) is an important cause of disease in older adults and is associated with high morbidity and mortality, especially in those with high-risk conditions. Illness can vary from mild upper respiratory tract symptoms to more severe lower respiratory tract disease. After over 50 years of research, there is now hope for an RSV vaccine for any population, including older adults. An investigational bivalent RSV A and B, stabilized RSV prefusion F subunit vaccine (RSVpreF) was assessed successfully in a pivotal phase 3 efficacy study in older adults. (NCT05035212). Methods The primary efficacy objective of this Phase 3, global, multicenter, randomized, double-blinded, placebo-controlled study was to evaluate the prevention of RSV associated lower respiratory tract illness (LRTI-RSV) in up to 40,000 adults ≥60 years of age during the first winter season (September 2021-June 2022). Two primary endpoints were tested sequentially – LRTI-RSV with ≥2 and ≥3 symptoms. A pre-planned efficacy interim analysis (IA) was to be conducted by an external Data Monitoring Committee (DMC) upon accrual of at least 29 cases of LRTI-RSV with ≥2 symptoms. With efficacy demonstrated for cases with ≥2 symptoms and sufficient cases with ≥ 3 symptoms accrued, an efficacy analysis of cases with ≥ 3 symptoms was to be conducted. The ongoing study is collecting additional safety and descriptive efficacy data. Results At the time of the IA, approximately 34,000 participants received either RSVpreF 120 µg (60 µg each of RSVpreF from RSV A and RSV B) or placebo (1:1 randomization). Forty-four LRTI-RSV cases with ≥2 symptoms were accrued with 11 cases in the RSVpreF group and 33 cases in the placebo group corresponding to a VE of 66.7% (96.66% CI: 28.8%, 85.8%). Sixteen LRTI-RSV cases with ≥3 symptoms were accrued with 2 cases in the RSVpreF group and 14 cases in the placebo group corresponding to a VE of 85.7% (96.66% CI: 32.0%, 98.7%). The investigational vaccine was well-tolerated with no safety concerns. Conclusion Despite unpredictable RSV activity due to the COVID-19 pandemic, the primary objective of the study was met demonstrating that RSVpreF had a favorable safety profile and was highly efficacious in preventing LRTI-RSV with ≥2 symptoms and ≥3 symptoms in older adults 60 years and older. Disclosures Edward E. Walsh, MD, Janssen: Honoraria|Merck: Grant/Research Support|Pfizer: Grant/Research Support Fernando Polack, MD, I-trials: Ownership Interest|Pfizer: Grant/Research Support Agnieszka Zareba, MD PhD, Pfizer: employee of Pfizer Ann R. Falsey, MD, BioFire Diagnostics: Grant/Research Support|Janssen: Grant/Research Support|Merck Sharp & Dohme: Grant/Research Support|Novavax: Advisor/Consultant|Pfizer: Grant/Research Support Gonzalo Perez Marc, MD, I-trials: Board Member|I-trials: Ownership Interest|Pfizer: Grant/Research Support Qin Jiang, PhD, Pfizer: employee of Pfizer Kathy Schneider, PhD, Pfizer: employee of Pfizer David Cooper, PhD, Pfizer: employee of Pfizer Maria Maddalena Lino, PhD, Pfizer: employee of Pfizer Annaliesa S. Anderson, PhD, Pfizer: employee of Pfizer|Pfizer Inc: Employee|Pfizer Inc: Stocks/Bonds Katherin U Jensen, PhD, Pfizer: employee of Pfizer Kena A. Swanson, Ph.D., Pfizer: employee of Pfizer Alejandra C. Gurtman, M.D., Pfizer: employee of Pfizer William C. Gruber, MD, Pfizer Inc: Employee|Pfizer Inc: Stocks/Bonds Beate Schmoele-Thoma, MD, Pfizer: employee of Pfizer.
- Published
- 2022
13. 2134. A Phase 2 Study to Evaluate the Safety, Tolerability, and Immunogenicity of a Booster Dose of a Group B Streptococcus 6-Valent Polysaccharide Conjugate Vaccine (GBS6)
- Author
-
Babalwa Jongihlati, Nathan Segall, Stanley Block, James Peterson, Judith Absalon, Samantha Munson, Yasmin Sanchez-Pearson, Raphael Simon, Natalie Silmon de Monerri, David Radley, Emily A Gomme, Michelle Gaylord, William C Gruber, Kathrin U Jansen, Daniel A Scott, and Annaliesa S Anderson
- Subjects
Infectious Diseases ,Oncology - Abstract
Background Group B streptococcus (GBS) is a leading cause of invasive bacterial infections in young infants and pregnant women. Pfizer is developing a hexavalent GBS vaccine (GBS6) as a maternal vaccine to prevent invasive GBS disease due to the 6 most prevalent serotypes in young infants. We previously reported Phase 1 safety and immunogenicity data for GBS6. There is precedent for repeat doses of vaccines to augment or sustain circulating antibodies available for placental transfer with each pregnancy; thus, data to inform GBS6 booster strategies were required. Methods This was a Phase 2 open-label extension study to evaluate the safety and immunogenicity of a booster dose of GBS6 (20 μg capsular polysaccharide (CPS)/serotype/dose) with or without AlPO4, given ∼2 years after primary vaccination in 151 healthy nonpregnant adults. Sera taken before and 1-month postbooster were assessed for anti-CPS IgG using a direct Luminex immunoassay. Participants recorded solicited local and systemic events for 14 days after vaccination and unsolicited safety events through 6 months after vaccination. Immunogenicity time points from the Phase 1 study are referred to as primary dose. Results Immunogenicity results For all serotypes, serotype-specific IgG geometric mean concentrations (GMCs) remained elevated compared to baseline at the prebooster time point and were higher 1-month postbooster than 1 month post primary. The 1-month postbooster IgG GMCs were 10- to 59-fold higher than at the prebooster time point. There were similar responses between the 2 formulations. Safety and tolerability results The most frequently reported local reaction was mild to moderate pain at the injection site. Greater pain was associated with AlPO4 formulation. The most frequently reported systemic events were mild to moderate headache and fatigue. The frequency of adverse events was low. Figure 1Antibody Response Line Plot of IgG GMCs by Vaccine Group - All SerotypesFigure 2.Reverse Cumulative Distribution Curves (RCDCs) for IgG 1 Month After Primary and Booster Vaccination, by Vaccine Group – All SerotypesFigure 3.IgG Geometric Mean Fold Rises (GMFRs) at 1 Month Postbooster Vaccination Conclusion A booster dose of GBS6 given ∼2 years after a primary dose to healthy nonpregnant adults was safe and elicited robust immune responses that were also consistently higher than after primary dose. This study suggests that repeat vaccination with GBS6 may confer additional benefit in pregnant women in subsequent pregnancies. Disclosures Babalwa Jongihlati, MD, MBA, Pfizer: Stocks/Bonds Judith Absalon, MD, MPH, Pfizer: Stocks/Bonds Samantha Munson, MPH, MBA, Pfizer: Stocks/Bonds Yasmin Sanchez-Pearson, PhD, Pfizer: Stocks/Bonds Raphael Simon, PhD, Pfizer: Stocks/Bonds Natalie Silmon de Monerri, PhD, Pfizer: Stocks/Bonds David Radley, MS, Pfizer: Employee|Pfizer: Stocks/Bonds Emily A. Gomme, Ph.D., Pfizer: Stocks/Bonds Michelle Gaylord, PhD, Pfizer: Stocks/Bonds William C. Gruber, MD, Pfizer, Inc.: Salary|Pfizer, Inc.: Stocks/Bonds Kathrin U. Jansen, PhD, Pfizer: Stocks/Bonds Daniel A. Scott, MD, Pfizer: Employee|Pfizer: Stocks/Bonds Annaliesa S. Anderson, PhD, Pfizer: Stocks/Bonds.
- Published
- 2022
14. Maternal immunization with Group B <scp> Streptococcus six‐valent </scp> polysaccharide conjugate vaccine supported by lack of toxicity in rat and rabbit fertility and developmental toxicity studies
- Author
-
Annaliesa S. Anderson, William S. Nowland, Natasha R. Catlin, Gregg D. Cappon, Scott Engel, Ingrid L. Scully, Christopher J. Bowman, Cynthia M. Rohde, and Sandra M. Buitrago
- Subjects
Embryology ,biology ,business.industry ,Offspring ,Health, Toxicology and Mutagenesis ,Antibody titer ,Developmental toxicity ,Physiology ,Toxicology ,Serology ,medicine.anatomical_structure ,Conjugate vaccine ,Lactation ,Pediatrics, Perinatology and Child Health ,biology.protein ,Medicine ,Gestation ,Antibody ,business ,Developmental Biology - Abstract
A maternal Group B Streptococcus (GBS) six-valent polysaccharide conjugate vaccine (GBS6) is being developed to protect neonates and infants up to 3 months of age through passive transfer of antibodies from the mother to the infant. Fertility and developmental toxicity studies were conducted in female Sprague Dawley rats and New Zealand White rabbits with GBS6 (20 μg capsular polysaccharide/serotype formulated with or without AlPO4 , the highest clinical dose). Females were administered the full human dose of the GBS6 formulation intramuscularly twice prior to mating and twice during gestation, to ensure that high antibody levels were maintained throughout gestation and lactation. Approximately, half of the rats and rabbits were evaluated at the end of gestation, and the remainder were evaluated at the end of lactation. Maternal blood for GBS6 serology, to measure antibody titers to the GBS6 antigens, was collected prior to the first dose, prior to mating, and at each necropsy. Blood for serology was also collected from offspring at the end of gestation and lactation. In both species, there was no evidence of vaccine-related effects on fertility, embryo-fetal development, or postnatal development of the offspring, supporting regulatory guidance that single-species evaluation would have been sufficient. Functional serum antibodies to all six serotypes in the vaccine were confirmed in maternal animals and functional serum antibodies to one or more of the six serotypes was also confirmed in some rat offspring and most of the rabbit offspring. The results of these studies supported the safety of GBS6 vaccine administration to pregnant women.
- Published
- 2021
15. Safety and immunogenicity of different Clostridioides (Clostridium) difficile vaccine formulations in two early phase randomized studies of healthy adults aged 50–85 years
- Author
-
Kathrin U. Jansen, Annaliesa S. Anderson, Yahong Peng, Charles Knirsch, Jody Lawrence, Kevin Yi, William C. Gruber, Chris Webber, Michael W. Pride, and Nicholas Kitchin
- Subjects
medicine.medical_specialty ,Placebo ,Immune system ,Clostridioides ,Internal medicine ,Humans ,Medicine ,Adverse effect ,Clostridium ,General Veterinary ,General Immunology and Microbiology ,biology ,Clostridioides difficile ,business.industry ,Immunogenicity ,Public Health, Environmental and Occupational Health ,Toxoid ,Toxoids ,Clostridium difficile ,Regimen ,Infectious Diseases ,Bacterial Vaccines ,biology.protein ,Molecular Medicine ,Antibody ,business - Abstract
Background Two phase 1/phase 2 studies assessed 2 formulations of investigational bivalent Clostridioides (Clostridium) difficile vaccine (QS-21 adjuvanted toxoid and toxoid-alone) in healthy adults 50–85 years of age. Methods The QS-21 adjuvanted toxoid vaccine study randomized subjects 3:1 to 100 μg QS-21–containing C difficile vaccine or placebo administered in a shortened-month (Months 0, 1, 3) or day (Days 1, 8, 30) regimen. The toxoid-alone vaccine study randomized subjects 3:3:1 to receive 100 or 200 μg unadjuvanted C difficile vaccine formulation or placebo in Stages 1 and 2 (sentinel cohorts of different age groups), and 3:1 to receive the selected dose of unadjuvanted C difficile vaccine formulation or placebo in Stage 3 (Days 1, 8, 30). Safety was the primary outcome for both studies. Immunogenicity was determined by measuring serum toxin A– and B–specific neutralizing antibodies. Results In the day regimen, 10 reports across both studies of grade 3 injection site redness postdose 2 triggered predefined stopping rules. Local reactions in both studies were more common among vaccine versus placebo recipients. Injection site pain predominated and was generally mild in severity. Systemic events were infrequent and generally mild-to-moderate in severity. Adverse events were reported by 50.0%–75.0% and 16.7%–50.0% of subjects in the QS-21 and toxoid-alone studies, respectively. Immune responses peaked around Day 37 (shortened-month regimen) or between Day 15 and Month 2 (day regimen) and remained above baseline throughout follow-up. Conclusions Both formulations demonstrated robust immunogenicity. Both studies stopped early due to grade 3 injection site redness postdose 2 of the day regimen; neither formulation progressed to later stage development. Instead, an aluminum hydroxide-containing formulation of the vaccine candidate administered at 0, 1, and 6 months, which was safe and immunogenic in phase 1 and 2 studies, advanced to phase 3 studies.
- Published
- 2021
16. Improved Neutralization of Omicron BA.4/5, BA.4.6, BA.2.75.2, BQ.1.1, and XBB.1 with Bivalent BA.4/5 Vaccine
- Author
-
Jing Zou, Chaitanya Kurhade, Sohil Patel, Nicholas Kitchin, Kristin Tompkins, Mark Cutler, David Cooper, Qi Yang, Hui Cai, Alexander Muik, Ying Zhang, Dung-Yang Lee, Ugur Sahin, Annaliesa S. Anderson, William C. Gruber, Xuping Xie, Kena A. Swanson, and Pei-Yong Shi
- Abstract
The BNT162b2 bivalent BA.4/5 COVID-19 vaccine has been authorized to mitigate COVID-19 due to current Omicron and potentially future variants. New sublineages of SARS-CoV-2 Omicron continue to emerge and have acquired additional mutations, particularly in the spike protein, that may lead to improved viral fitness and immune evasion. The present study characterized neutralization activities against new Omicron sublineages BA.4.6, BA.2.75.2, BQ.1.1, and XBB.1 after a 4thdose (following three doses of BNT162b2) of either the original monovalent BNT162b2 or the bivalent BA.4/5 booster in individuals >55 years of age. For all participants, the 4thdose of monovalent BNT162b2 vaccine induced a 3.0×, 2.9×, 2.3×, 2.1×, 1.8×, and 1.5× geometric mean neutralizing titer fold rise (GMFR) against USA/WA1-2020 (a strain isolated in January 2020), BA.4/5, BA.4.6, BA.2.75.2, BQ.1.1, and XBB.1, respectively; the bivalent vaccine induced 5.8×, 13.0×, 11.1×, 6.7×, 8.7×, and 4.8× GMFRs. For individuals without SARS-CoV-2 infection history, BNT162b2 monovalent induced 4.4×, 3.0×, 2.5×, 2.0×, 1.5×, and 1.3× GMFRs, respectively; the bivalent vaccine induced 9.9×, 26.4×, 22.2×, 8.4×, 12.6×, and 4.7× GMFRs. These data suggest the bivalent BA.4/5 vaccine is more immunogenic than the original BNT162b2 monovalent vaccine against circulating Omicron sublineages, including BQ.1.1 that is becoming prevalent globally.
- Published
- 2022
17. Genetic Surveillance of SARS-CoV-2 M pro Reveals High Sequence and Structural Conservation Prior to the Introduction of Protease Inhibitor Paxlovid
- Author
-
Jonathan T. Lee, Qingyi Yang, Alexey Gribenko, B. Scott Perrin, Yuao Zhu, Rhonda Cardin, Paul A. Liberator, Annaliesa S. Anderson, and Li Hao
- Subjects
Virology ,Microbiology - Abstract
The recent authorization of oral severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antivirals, such as Paxlovid, has ushered in a new era of the COVID-19 pandemic. The emergence of new variants, as well as the selective pressure imposed by antiviral drugs themselves, raises concern for potential escape mutations in key drug binding motifs.
- Published
- 2022
18. Genetic Surveillance of SARS-CoV-2 M
- Author
-
Jonathan T, Lee, Qingyi, Yang, Alexey, Gribenko, B Scott, Perrin, Yuao, Zhu, Rhonda, Cardin, Paul A, Liberator, Annaliesa S, Anderson, and Li, Hao
- Subjects
Ritonavir ,Lactams ,Proline ,SARS-CoV-2 ,COVID-19 ,Viral Nonstructural Proteins ,Antiviral Agents ,Cysteine Endopeptidases ,Drug Combinations ,Leucine ,Nitriles ,Animals ,Humans ,Protease Inhibitors ,Pandemics ,Coronavirus 3C Proteases - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to represent a global health emergency as a highly transmissible, airborne virus. An important coronaviral drug target for treatment of COVID-19 is the conserved main protease (M
- Published
- 2022
19. Semisynthetic Glycoconjugate Vaccine Candidates against
- Author
-
Arun, Naini, Max Peter, Bartetzko, Someswara Rao, Sanapala, Felix, Broecker, Victoria, Wirtz, Marilda P, Lisboa, Sharavathi G, Parameswarappa, Daniel, Knopp, Jessica, Przygodda, Matthias, Hakelberg, Rosalind, Pan, Axay, Patel, Laurent, Chorro, Arthur, Illenberger, Christopher, Ponce, Srinivas, Kodali, Jacqueline, Lypowy, Annaliesa S, Anderson, Robert G K, Donald, Arne, von Bonin, and Claney L, Pereira
- Abstract
Extraintestinal pathogenic
- Published
- 2022
20. Human Infection Challenge with Serotype 3 Pneumococcus
- Author
-
Ryan E. Robinson, Elena Mitsi, Elissavet Nikolaou, Sherin Pojar, Tao Chen, Jesús Reiné, Tinashe K. Nyazika, James Court, Kelly Davies, Madlen Farrar, Patricia Gonzalez-Dias, Josh Hamilton, Helen Hill, Lisa Hitchins, Ashleigh Howard, Angela Hyder-Wright, Maia Lesosky, Konstantinos Liatsikos, Agnes Matope, Daniella McLenaghan, Christopher Myerscough, Annabel Murphy, Carla Solórzano, Duolao Wang, Hassan Burhan, Manish Gautam, Elizabeth Begier, Christian Theilacker, Rohini Beavon, Annaliesa S. Anderson, Bradford D. Gessner, Stephen B. Gordon, Andrea M. Collins, and Daniela M. Ferreira
- Subjects
Pulmonary and Respiratory Medicine ,Adult ,wc_20 ,Infant ,wc_204 ,Critical Care and Intensive Care Medicine ,Serogroup ,Pneumococcal Infections ,wc_200 ,Anti-Bacterial Agents ,Pneumococcal Vaccines ,Young Adult ,Streptococcus pneumoniae ,Nasopharynx ,Carrier State ,Humans ,Child - Abstract
RationaleStreptococcus pneumoniae serotype 3 (SPN3) is a cause of invasive pneumococcal disease and associated with low carriage rates. Following the introduction of pediatric 13-valent pneumococcal conjugate vaccine (PCV13) programmes, SPN3 declines are less than other vaccine serotypes and incidence has increased in some populations coincident with a shift in predominant circulating SPN3 clade, from I to II. A human challenge model provides an effective means for assessing the impact of PCV13 on SPN3 in the upper airway.ObjectivesTo establish SPN3's ability to colonise the nasopharynx using different inoculum clades and doses and the safety of an SPN3 challenge model.MethodsIn a human challenge study involving three well characterised and antibiotic sensitive SPN3 isolates (PFESP306 [clade Ia], PFESP231 [no clade] and PFESP505 [clade II]), inoculum doses (10,000, 20,000, 80,000, 160,000 CFU/100μL) were escalated until maximal colonisation rates were achieved, with concurrent acceptable safety.Outcome measuresPresence and density of experimental SPN3 nasopharyngeal colonisation in nasal wash samples, assessed using microbiological culture and molecular methods, on days 2, 7 and 14 post-inoculation.Results96 healthy participants (median age 21, interquartile range 19-25) were inoculated (n=6-10 per dose group, 10 groups). Colonisation rates ranged from 30.0-70.0% varying with dose and isolate. 30.0% (29/96) reported mild symptoms (82.8% sore throat, [24/29]), one developed otitis media requiring antibiotics. No serious adverse events occurred.ConclusionsAn SPN3 human challenge model is feasible and safe with comparable carriage rates to an established SPN6B human challenge model. SPN3 carriage may cause mild upper respiratory symptoms. Clinical trial registration available at www. https://www.isrctn.com/, ID: ISRCTN11306486.
- Published
- 2022
21. Protocol for a phase IV double-blind randomised controlled trial to investigate the effect of the 13-valent pneumococcal conjugate vaccine and the 23-valent pneumococcal polysaccharide vaccine on pneumococcal colonisation using the experimental human pneumococcal challenge model in healthy adults (PREVENTING PNEUMO 2)
- Author
-
Konstantinos, Liatsikos, Angela, Hyder-Wright, Sherin, Pojar, Tao, Chen, Duolao, Wang, Kelly, Davies, Christopher, Myerscough, Jesus, Reine, Ryan E, Robinson, Britta, Urban, Elena, Mitsi, Carla, Solorzano, Stephen B, Gordon, Angela, Quinn, Kaijie, Pan, Annaliesa S, Anderson, Christian, Theilacker, Elizabeth, Begier, Bradford D, Gessner, Andrea, Collins, Daniela M, Ferreira, and Ben, Morton
- Subjects
Adult ,Vaccines, Conjugate ,Adolescent ,General Medicine ,Clinical Trials, Phase IV as Topic ,Middle Aged ,Serogroup ,Pneumococcal Infections ,Pneumococcal Vaccines ,Otitis Media ,Young Adult ,Streptococcus pneumoniae ,Humans ,Randomized Controlled Trials as Topic - Abstract
IntroductionDespite widely available vaccinations, Streptococcus pneumoniae (SPN) remains a major cause of morbidity and mortality worldwide, causing community-acquired pneumonia, meningitis, otitis media, sinusitis and bacteraemia. Here, we summarise an ethically approved protocol for a double-blind, randomised controlled trial investigating the effect of the 13-valent pneumococcal conjugate vaccine (PCV13) and the 23-valent pneumococcal polysaccharide vaccine (PPV23) on pneumococcal nasopharyngeal colonisation acquisition, density and duration using experimental human pneumococcal challenge (EHPC).Methods and analysisHealthy adult participants aged 18–50 years will be randomised to receive PCV13, PPV23 or placebo and then undergo one or two EHPCs involving intranasal administration of SPN at 1-month post-vaccination with serotype 3 (SPN3) and 6 months with serotype 6B (SPN6B). Participants randomised to PCV13 and placebo will also be randomised to one of two clinically relevant SPN3 strains from distinct lineages within clonal complex 180, clades Ia and II, creating five study groups. Following inoculation, participants will be seen on days 2, 7, 14 and 23. During the follow-up period, we will monitor safety, colonisation status, density and duration, immune responses and antigenuria. The primary outcome of the study is comparing the rate of SPN3 acquisition between the vaccinated (PCV13 or PPV23) and unvaccinated (placebo) groups as defined by classical culture. Density and duration of colonisation, comparison of acquisition rates using molecular methods and evaluation of the above measurements for individual SPN3 clades and SPN6B form the secondary objectives. Furthermore, we will explore the immune responses associated with these vaccines, their effect on colonisation and the relationship between colonisation and urinary pneumococcal antigen detection.Ethics and disseminationThe study is approved by the NHS Research and Ethics Committee (Reference: 20/NW/0097) and by the Medicines and Healthcare products Regulatory Agency (Reference: CTA 25753/0001/001–0001). Findings will be published in peer-reviewed journals.Trial registration numberISRCTN15728847, NCT04974294.
- Published
- 2022
22. Safety and immunogenicity of a novel hexavalent group B streptococcus conjugate vaccine in healthy, non-pregnant adults: a phase 1/2, randomised, placebo-controlled, observer-blinded, dose-escalation trial
- Author
-
Judith Absalon, Nathan Segall, Stan L Block, Kimberly J Center, Ingrid L Scully, Peter C Giardina, James Peterson, Wendy J Watson, William C Gruber, Kathrin U Jansen, Yahong Peng, Samantha Munson, Danka Pavliakova, Daniel A Scott, and Annaliesa S Anderson
- Subjects
0301 basic medicine ,medicine.medical_specialty ,education.field_of_study ,business.industry ,030106 microbiology ,Population ,Placebo ,Group B ,law.invention ,Clinical trial ,Vaccination ,03 medical and health sciences ,0302 clinical medicine ,Infectious Diseases ,Randomized controlled trial ,law ,Internal medicine ,Medicine ,030212 general & internal medicine ,Intramuscular injection ,business ,education ,Adverse effect - Abstract
Summary Background Group B streptococcus (GBS) is a major cause of invasive disease in young infants. Infants born to women with sufficient pre-existing anti-GBS capsular IgG antibodies are at reduced risk of GBS disease, making maternal immunisation a potential strategy for prevention. We aimed to assess the safety and immunogenicity of a novel hexavalent (serotypes Ia, Ib, II, III, IV, and V) GBS conjugate vaccine (GBS6). Methods This phase 1/2, placebo-controlled, observer-blinded, dose-escalation trial, was done at four clinical research centres in the USA (Kentucky, Georgia, and two sites in Utah). Healthy, non-pregnant adults aged 18–49 years were randomly assigned using an interactive, web-based response technology system. Within each dose group (low, medium, or high), participants in sentinel cohorts were randomly assigned 2:2:1 and expanded cohort participants were randomly assigned 4:4:1 to receive GBS6 with aluminium phosphate (AlPO4), GBS6 without AlPO4, or placebo (saline control). One 0·5 mL dose of either saline placebo or 5 μg capsular polysaccharide per serotype in the low-dose group, 10 μg capsular polysaccharide per serotype in the medium-dose group, or 20 μg capsular polysaccharide per serotype in the high-dose group was administered by intramuscular injection into the deltoid muscle on day 1. The primary outcome was safety up to 6 months after vaccination, including the proportion of sentinel cohort participants with clinical laboratory abnormalities at 1 week, the proportion of all participants reporting solicited local reactions, systemic events, or use of antipyretic or pain medication within 14 days, adverse events up to 1 month, and medically attended or serious adverse events up to 6 months. The secondary outcome was GBS immunogenicity (serotype-specific IgG geometric mean concentrations at 1 month). This study is registered with ClinicalTrials.gov , NCT03170609 . Findings Between June 5, 2017, and June 25, 2018, 365 participants were randomly assigned and 364 (52 in each dose group) were vaccinated and included in the safety analysis. Unsolicited adverse events were reported by 15 (29%) participants in the 5 μg with AlPO4 group, 13 (25%) in the 5 μg without AlPO4 group, 22 (42%) in the 10 μg with AlPO4 group, 12 (23%) in the 10 μg without AlPO4 group, 25 (48%) in the 20 μg with AlPO4 group, 21 (40%) in the 20 μg without AlPO4 group, and 20 (38%) in the placebo group. The most common unsolicited adverse events were in the system organ class of infections and infestations in any dose or formulation of GBS6 (ranging from six [12%] in the 10 μg without AlPO4 group to 15 [29%] in the 20 μg with AlPO4 group and placebo group). Three participants reported at least one serious adverse event during the study, one each in the 5 μg GBS6 with AlPO4 group (diabetic ketoacidosis, two events; resolved), 10 μg GBS6 with AlPO4 group (died by suicide), and 20 μg GBS6 with AlPO4 group (metrorrhagia; resolved). None of these serious adverse events were considered related to the vaccine. 11 of the 365 participants were excluded from the evaluable immunogenicity population, including one participant who did not receive the vaccine, and ten who at 1 month after vaccination were withdrawn for various reasons. GBS serotype-specific IgG geometric mean concentrations increased by 1 week after vaccination for all GBS6 groups, peaked at 2 weeks, stabilised by 1 month, and declined gradually but remained higher than placebo at 6 months. Interpretation GBS6 was well tolerated in healthy adults and elicited robust immune responses for all dose levels and formulations that persisted 6 months after vaccination. This study supports further evaluation of GBS6 in pregnant women. Funding Pfizer.
- Published
- 2021
23. Correlates of protection for meningococcal surface protein vaccines: current approaches for the determination of breadth of coverage
- Author
-
Jamie Findlow, Ray Borrow, David S. Stephens, Paul Liberator, Annaliesa S. Anderson, Paul Balmer, and Luis Jodar
- Subjects
Pharmacology ,Meningococcal Infections ,Antigens, Bacterial ,Drug Discovery ,Immunology ,Bacterial Vaccines ,Molecular Medicine ,Humans ,Membrane Proteins ,Meningococcal Vaccines ,Neisseria meningitidis ,Neisseria meningitidis, Serogroup B - Abstract
The two currently licensed surface protein non-capsular meningococcal serogroup B (MenB) vaccines both have the purpose of providing broad coverage against diverse MenB strains. However, the different antigen compositions and approaches used to assess breadth of coverage currently make direct comparisons complex.In the second of two companion papers, we comprehensively review the serology and factors influencing breadth of coverage assessments for two currently licensed MenB vaccines.Surface protein MenB vaccines were developed using different approaches, resulting in unique formulations and thus their breadth of coverage. The surface proteins used as vaccine antigens can vary among meningococcal strains due to gene presence/absence, sequence diversity, and differences in protein expression. Assessment of the breadth of coverage provided by vaccines is influenced by the ability to induce cross-reactive functional immune responses to sequence diverse protein variants; the characteristics of the circulating invasive strains from specific geographic locations; methodological differences in the immunogenicity assays; differences in human immune responses between individuals; and the maintenance of protective antibody levels over time. Understanding the proportion of meningococcal strains, which are covered by the two licensed vaccines, is important in understanding protection from disease and public health use.
- Published
- 2022
24. Genetic surveillance of SARS-CoV-2 Mpro reveals high sequence and structural conservation prior to the introduction of protease inhibitor Paxlovid
- Author
-
Jonathan T. Lee, Qingyi Yang, Alexey Gribenko, B. Scott Perrin, Yuao Zhu, Rhonda Cardin, Paul A. Liberator, Annaliesa S. Anderson, and Li Hao
- Abstract
SARS-CoV-2 continues to represent a global health emergency as a highly transmissible, airborne virus. An important coronaviral drug target for treatment of COVID-19 is the conserved main protease (Mpro). Nirmatrelvir is a potent Mpro inhibitor and the antiviral component of Paxlovid™. The significant viral sequencing effort during the ongoing COVID-19 pandemic represented a unique opportunity to assess potential nirmatrelvir escape mutations from emerging variants of SARS-CoV-2. To establish the baseline mutational landscape of Mpro prior to the introduction of Mpro inhibitors, Mpro sequences and its cleavage junction regions were retrieved from ∼4,892,000 high-quality SARS-CoV-2 genomes in GISAID. Any mutations identified from comparison to the reference sequence (Wuhan-hu-1) were cataloged and analyzed. Mutations at sites key to nirmatrelvir binding and protease functionality (e.g., dimerization sites) were still rare. Structural comparison of Mpro also showed conservation of key nirmatrelvir contact residues across the extended Coronaviridae family (alpha-, beta-, and gamma-coronaviruses). Additionally, we showed that over time the SARS-CoV-2 Mpro enzyme remained under purifying selection and was highly conserved relative to the spike protein. Now, with the EUA approval of Paxlovid and its expected widespread use across the globe, it is essential to continue large-scale genomic surveillance of SARS-CoV-2 Mpro evolution. This study establishes a robust analysis framework for monitoring emergent mutations in millions of virus isolates, with the goal of identifying potential resistance to present and future SARS-CoV-2 antivirals.ImportanceThe recent authorization of oral SARS-CoV-2 antivirals, such as Paxlovid, has ushered in a new era of the COVID-19 pandemic. Emergence of new variants, as well as selective pressure imposed by antiviral drugs themselves, raise concern for potential escape mutations in key drug binding motifs. To determine the potential emergence of antiviral resistance in globally circulating isolates and its implications for the clinical response to the COVID-19 pandemic, sequencing of SARS-CoV-2 viral isolates before, during, and after the introduction of new antiviral treatments is critical. The infrastructure built herein for active genetic surveillance of Mpro evolution and emergent mutations will play an important role in assessing potential antiviral resistance as the pandemic progresses and Mpro inhibitors are introduced. We anticipate our framework to be the starting point in a larger effort for global monitoring of the SARS-CoV-2 Mpro mutational landscape.
- Published
- 2022
25. Preclinical Immunogenicity and Efficacy of Optimized O25b O-Antigen Glycoconjugates To Prevent MDR ST131 E. coli Infections
- Author
-
Laurent Chorro, Zhenghui Li, Ling Chu, Suddham Singh, Jianxin Gu, Jin-hwan Kim, Kaushik Dutta, Rosalind Pan, Srinivas Kodali, Duston Ndreu, Axay Patel, Julio C. Hawkins, Chris Ponce, Natalie Silmon de Monerri, David Keeney, Arthur Illenberger, C. Hal Jones, Lubomira Andrew, Jason Lotvin, A. Krishna Prasad, Isis Kanevsky, Kathrin U. Jansen, Annaliesa S. Anderson, and Robert G. K. Donald
- Subjects
Mice ,Infectious Diseases ,Immunology ,Escherichia coli ,Animals ,O Antigens ,Parasitology ,Carrier Proteins ,Microbiology ,Glycoconjugates ,Escherichia coli Infections - Abstract
Multivalent O-antigen polysaccharide glycoconjugate vaccines are under development to prevent invasive infections caused by pathogenic Enterobacteriaceae . Sequence type 131 (ST131) Escherichia coli of serotype O25b has emerged as the predominant lineage causing invasive multidrug-resistant extraintestinal pathogenic E. coli (ExPEC) infections. We observed the prevalence of E. coli O25b ST131 among a contemporary collection of isolates from U.S. bloodstream infections from 2013 to 2016 ( n = 444) and global urinary tract infections from 2014 to 2017 ( n = 102) to be 25% and 24%, respectively.
- Published
- 2022
26. Advances towards licensure of a maternal vaccine for the prevention of invasive group B streptococcus disease in infants: a discussion of different approaches
- Author
-
Judith Absalon, Raphael Simon, David Radley, Peter C. Giardina, Kenneth Koury, Kathrin U. Jansen, and Annaliesa S. Anderson
- Subjects
Pharmacology ,Pregnancy ,Streptococcal Infections ,Immunology ,Streptococcal Vaccines ,Infant, Newborn ,Immunology and Allergy ,Humans ,Infant ,Female ,Pregnancy Complications, Infectious ,Infectious Disease Transmission, Vertical ,Streptococcus agalactiae - Abstract
Group B streptococcus (
- Published
- 2022
27. Accelerated Preclinical Paths to Support Rapid Development of COVID-19 Therapeutics
- Author
-
Jay A. Grobler, Christine M. Colvis, Rosa M. Alvarez, John A. T. Young, Michael S. Diamond, Kara Carter, Joseph P. Menetski, Prabhavathi Fernandes, and Annaliesa S. Anderson
- Subjects
Prioritization ,0303 health sciences ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Drug discovery ,Process (engineering) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Biology ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Risk analysis (engineering) ,Virology ,Parasitology ,030217 neurology & neurosurgery ,Coronavirus Infections ,030304 developmental biology - Abstract
When SARS-CoV-2 emerged at the end of 2019, no approved therapeutics or vaccines were available. An urgent need for countermeasures during this crisis challenges the current paradigm of traditional drug discovery and development, which usually takes years from start to finish. Approaches that accelerate this process need to be considered. Here we propose the minimum data package required to move a compound into clinical development safely. We further define the additional data that should be collected in parallel without impacting the rapid path to clinical development. Accelerated paths for antivirals, immunomodulators, anticoagulants, and other agents have been developed and can serve as "roadmaps" to support prioritization of compounds for clinical testing. These accelerated paths are fueled by a skewed risk-benefit ratio and are necessary to advance therapeutic agents into human trials rapidly and safely for COVID-19. Such paths are adaptable to other potential future pandemics.
- Published
- 2020
28. MenB-FHbp Vaccine Protects Against Diverse Meningococcal Strains in Adolescents and Young Adults: Post Hoc Analysis of Two Phase 3 Studies
- Author
-
Jean-Louis Pregaldien, Paul Balmer, Joseph Eiden, Robert E. O’Neill, Johannes Beeslaar, David Radley, Judith Absalon, Thomas R. Jones, Roger Maansson, Shannon L. Harris, Amit Srivastava, Annaliesa S. Anderson, John L. Perez, and John Ginis
- Subjects
0301 basic medicine ,Microbiology (medical) ,030106 microbiology ,Infectious and parasitic diseases ,RC109-216 ,Neisseria meningitidis ,Bactericidal activity ,Meningococcal serogroup B ,Meningococcal disease ,medicine.disease_cause ,Adolescents ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Post-hoc analysis ,Medicine ,030212 general & internal medicine ,Young adult ,Original Research ,Broad coverage ,business.industry ,FHbp ,medicine.disease ,MenB-FHbp vaccine ,Hepatitis A virus vaccine ,Vaccination ,Titer ,hSBA ,Infectious Diseases ,Immunology ,business ,Young adults - Abstract
Introduction Two phase 3 studies in adolescents and young adults demonstrated that MenB-FHbp, a meningococcal serogroup B (MenB) vaccine, elicits protective immune responses after 2 or 3 doses based on serum bactericidal antibody assays using human complement (hSBA) against 4 primary and 10 additional diverse, vaccine-heterologous MenB test strains. Lower limits of quantitation (LLOQs; titers 1:8 or 1:16; titers ≥ 1:4 correlate with protection) were used to evaluate responses to individual strains and all 4 primary strains combined (composite response). A post hoc analysis evaluated percentages of subjects with protective responses to as many as 8 strains combined (4 primary plus additional strains). Methods Immune responses were measured using hSBAs against 4 primary strains in adolescents (n = 1509, MenB-FHbp; n = 898, hepatitis A virus vaccine/saline) and young adults (n = 2480, MenB-FHbp; n = 824, saline) receiving MenB-FHbp or control at 0, 2, and 6 months. Ten additional strains were evaluated in subsets of subjects from approximately 1800 MenB-FHbp recipients across both studies. Percentages of subjects with hSBA titers ≥ LLOQ for different numbers of primary strains or primary plus additional strains combined (7 or 8 strains total per subset) were determined before vaccination, 1 month post-dose 2, and 1 month post-dose 3. Results Across the panel of primary plus additional strains, at 1 month post-dose 3, titers ≥ LLOQ were elicited in 93.7–95.7% of adolescents and 91.7–95.0% of young adults for ≥ 5 test strains combined and in 70.5–85.8% of adolescents and 67.5–81.4% of young adults for ≥ 7 strains combined. Among adolescents, 99.8%, 99.0%, 92.8%, and 82.7% had titers ≥ LLOQ against at least 1, 2, 3, and all 4 primary strains, respectively; corresponding percentages for young adults were 99.7%, 97.7%, 94.0%, and 84.5%. Conclusions Results support the ability of MenB-FHbp to provide broad coverage against MenB strains expressing diverse FHbp variants. Trial Registration ClinicalTrials.gov identifiers NCT01830855, NCT01352845. Electronic supplementary material The online version of this article (10.1007/s40121-020-00319-0) contains supplementary material, which is available to authorized users.
- Published
- 2020
29. Peripheral CD4 T follicular cells induced by a conjugated pneumococcal vaccine correlate with enhanced opsonophagocytic antibody responses in younger individuals
- Author
-
Harry W. Schroeder, Robert L. Burton, A. Krishna Prasad, Andrew O. Westfall, Anju Bansal, Annaliesa S. Anderson, Suddham Singh, Gregory C. Ippolito, Michael W. Pride, Binghao J. Peng, Sarah Sterrett, Paul A. Goepfert, David C. LaFon, and Moon H. Nahm
- Subjects
CD4-Positive T-Lymphocytes ,Serotype ,030231 tropical medicine ,Immunization, Secondary ,Pneumococcal Infections ,Article ,Flow cytometry ,Pneumococcal Vaccines ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Phagocytosis ,Follicular phase ,Humans ,Medicine ,030212 general & internal medicine ,Vaccines, Conjugate ,General Veterinary ,General Immunology and Microbiology ,medicine.diagnostic_test ,biology ,business.industry ,Age Factors ,Public Health, Environmental and Occupational Health ,T-Lymphocytes, Helper-Inducer ,Antibodies, Bacterial ,Vaccination ,Titer ,Infectious Diseases ,Pneumococcal vaccine ,Antibody Formation ,Immunology ,biology.protein ,Molecular Medicine ,Antibody ,business - Abstract
Background PCV13 (conjugated polysaccharide) and PPSV23 (polysaccharide only) are two licensed vaccines targeting S. pneumoniae. The role of CD4 T-cell responses in pneumococcal vaccines among healthy participants and their impact on antibodies is not yet known. Methods Ten adults (5 old and 5 young) received PCV13 (prime) and a year later PPSV23 (boost). Blood samples were collected prior to and multiple time points after vaccination. CD4 T cells responding to CRM197, polysaccharide (PS), CRM197 conjugated polysaccharide (CPS), PCV13 and PPSV23 vaccines were measured by flow cytometry. Serum antibodies were analyzed via multiplex opsonophagocytosis (MOPA) and pneumococcal IgG assays. Results Vaccine-specific CD4 T cells were induced in all ten vaccinees post PCV13. Older vaccinees mounted higher peak responses and those specific for PCV13 and conjugated PS-1 were more polyfunctional compared to the younger group. Vaccine-elicited peripheral T follicular helper (Tfh) cells were only detected in the younger group who also exhibited a higher fold change in OPA titers post both vaccines. Importantly, Tfh cells following PCV13 correlated only with PCV13 serotype specific OPA titers after PPSV23 vaccination. Conclusions These findings demonstrate age related differences in immune response and the potential importance of Tfh in modulating functional antibody responses following pneumococcal vaccination.
- Published
- 2020
30. Innovative Randomized Phase 1 Study and Dosing Regimen Selection to Accelerate and Inform Pivotal COVID-19 Trial of Nirmatrelvir
- Author
-
Ravi Shankar P. Singh, Sima S. Toussi, Frances Hackman, Phylinda L. Chan, Rohit Rao, Richard Allen, Lien Van Eyck, Sylvester Pawlak, Eugene P. Kadar, Frances Clark, Haihong Shi, Annaliesa S. Anderson, Michael Binks, Sandeep Menon, Gianluca Nucci, and Arthur Bergman
- Abstract
BackgroundCOVID-19 is a continued leading cause of hospitalization and death. Safe and efficacious COVID-19 antivirals are needed urgently. Nirmatrelvir (PF-07321332), the first orally bioavailable, SARS-CoV-2 Mpro inhibitor against the coronaviridae family, has demonstrated potent preclinical antiviral activity and benign safety profile.MethodsWe report safety, tolerability, and pharmacokinetic data of nirmatrelvir with and without ritonavir as a pharmacokinetic enhancer, from an accelerated randomized, double-blind, placebo-controlled, phase 1 study. Two interleaving single-ascending dose (SAD) cohorts were evaluated in a 3-period crossover. Multiple-ascending dose (MAD) with nirmatrelvir/ritonavir twice daily (BID) dosing was evaluated over 10 days in 5 parallel cohorts. Safety was assessed, including in a supratherapeutic exposure cohort. Dose and dosing regimen for clinical efficacy evaluation in phase 2/3 clinical trials were supported by integrating modelling and simulations of SAD/MAD data with nonclinical data and a quantitative systems pharmacology model (QSP).ResultsIn SAD, MAD, and supratherapeutic exposure cohorts, nirmatrelvir/ritonavir was safe and well tolerated. Nirmatrelvir exposure and half-life were considerably increased by ritonavir, enabling selection of nirmatrelvir/ritonavir dose and regimen for phase 2/3 trials (300/100 mg BID), to achieve concentrations continuously above those required for 90% inhibition of viral replication in vitro. The QSP model suggested that a 5-day regimen would significantly decrease viral load in SARS-CoV-2-infected patients and prevent development of severe disease, hospitalization, and death.ConclusionsAn innovative and seamless trial design expedited establishment of phase 1 safety and pharmacokinetics of nirmatrelvir/ritonavir, enabling high confidence in phase 2/3 dose selection and accelerated pivotal trials’ initiation. NCT04756531
- Published
- 2022
31. Nirmatrelvir, an orally active Mpro inhibitor, is a potent inhibitor of SARS-CoV-2 Variants of Concern
- Author
-
Devendra K. Rai, Irina Yurgelonis, Patricia McMonagle, Hussin A. Rothan, Li Hao, Alexey Gribenko, Elizabeth Titova, Barry Kreiswirth, Kris M. White, Yuao Zhu, Annaliesa S. Anderson, and Rhonda D. Cardin
- Abstract
New variants of SARS-CoV-2 with potential for enhanced transmission, replication, and immune evasion capabilities continue to emerge causing reduced vaccine efficacy and/or treatment failure. As of January 2021, the WHO has defined five ‘variants of concern’ (VOC): B.1.1.7 (Alpha, α), B.1.351 (Beta, β), P.1 (Gamma, γ), B.1.617.2 (Delta, δ), and B.1.1.529 (Omicron, o). To provide a therapeutic option for the treatment of COVID-19 and variants, Nirmatrelvir, the antiviral component of PAXLOVID™, an oral outpatient treatment recently authorized for conditional or emergency use treatment of COVID-19, was developed to inhibit SARS-CoV-2 replication. Nirmatrelvir (PF-07321332) is a specific inhibitor of coronavirus main protease (Mpro, also referred to as 3CLpro), with potent antiviral activity against several human coronaviruses, including SARS-CoV-2, SARS-CoV, and MERS (Owen et al, Science 2021. doi: 10.1126/science.abl4784). Here, we evaluated PF-07321332 against the five SARS-CoV-2 VOC (α, β, γ, δ,, o) and two Variants of Interest or VOI, C.37 (λ) and B.1.621 (μ), using qRT-PCR in VeroE6 cells lacking the P-glycoprotein (Pgp) multidrug transporter gene (VeroE6 P-gp knockout cells). Nirmatrelvir potently inhibited USA-WA1/2020 strain, and α, β, γ, λ, δ, μ, and o variants in VeroE6 P-gp knockout cells with mean EC50 values 38.0 nM, 41.0 nM, 127.2 nM, 24.9 nM, 21.2 nM, 15.9 nM, 25.7 nM and 16.2 nM, respectively. Sequence analysis of the Mpro encoded by the variants showed ~100% identity of active site amino acid sequences, reflecting the essential role of Mpro during viral replication leading to ability of Nirmatrelvir to exhibit potent activity across all the variants.
- Published
- 2022
32. S. aureus colonization in healthy Australian adults receiving an investigational S. aureus 3-antigen vaccine
- Author
-
Kathrin U. Jansen, Peter Richmond, Barry N. Kreiswirth, William C. Gruber, Michael D. Nissen, Helen Marshall, Joseph Eiden, Annaliesa S. Anderson, C. Hal Jones, Joseph M. Severs, Edward T. Zito, James Baber, and Sepehr Shakib
- Subjects
Adult ,0301 basic medicine ,Microbiology (medical) ,Staphylococcus aureus ,medicine.medical_specialty ,Adolescent ,030106 microbiology ,Oropharynx ,Booster dose ,Perineum ,medicine.disease_cause ,Placebo ,Placebos ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Double-Blind Method ,Antigen ,Internal medicine ,Prevalence ,Humans ,Medicine ,Colonization ,030212 general & internal medicine ,Immunization Schedule ,Aged ,Aged, 80 and over ,Antigens, Bacterial ,business.industry ,Australia ,Staphylococcal Vaccines ,Middle Aged ,Staphylococcal Infections ,Methicillin-resistant Staphylococcus aureus ,Healthy Volunteers ,Vaccination ,Nasal Mucosa ,Treatment Outcome ,Infectious Diseases ,Carriage ,Carrier State ,Vaccines, Subunit ,business - Abstract
Objectives Assess Staphylococcus aureus (S. aureus) colonization in healthy Australian adults receiving an investigational S. aureus 3-antigen vaccine (SA3Ag). Methods In this phase 1, double-blind, sponsor-unblinded study, participants were randomized to receive a single dose (1 of 3 dose levels) of SA3Ag or placebo and a booster dose or placebo at 6 months. S. aureus isolates from nasal, perineal, and oropharyngeal swabs before and through 12 months post-vaccination were identified. Results Baseline S. aureus colonization prevalence was 30.6% (any site), with nasal carriage (27.0%) more common than oropharyngeal/perineal (3.2% each). Following initial vaccination (low-dose: 102; mid-dose: 101; high-dose: 101; placebo: 102) and booster (low-dose: 45; mid-dose: 44; high-dose: 27; placebo: 181), placebo and SA3Ag groups showed similar S. aureus carriage through 12 months. Most colonized participants (74.0%) were colonized by single spa types. Placebo and SA3Ag groups had similar persistence of colonization, with 19.6–30.7% due to single spa types. Acquisition was observed in mid- and high-dose recipients (∼20%) and low-dose and placebo recipients (∼12%). Vaccination resulted in substantial increases in antibodies to all 3 antigens, irrespective of carriage status. Conclusions Based on descriptive analyses of this small study, SA3Ag vaccination did not impact S. aureus acquisition or carriage. Carriage status did not impact antibody responses to SA3Ag.
- Published
- 2019
33. Generation of a VeroE6 Pgp gene knock out cell line and its use in SARS-CoV-2 antiviral study
- Author
-
Yuao Zhu, Joe Binder, Irina Yurgelonis, Devendra K. Rai, Sarah Lazarro, Chester Costales, Keith Kobylarz, Patricia McMonagle, Claire M. Steppan, Lisa Aschenbrenner, Annaliesa S. Anderson, and Rhonda D. Cardin
- Subjects
Pharmacology ,Gene Knockout Techniques ,SARS-CoV-2 ,Virology ,Chlorocebus aethiops ,Humans ,Animals ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Antiviral Agents ,Vero Cells ,Cell Line ,COVID-19 Drug Treatment - Abstract
Vero cells are widely used for antiviral tests and virology research for SARS-CoV-2 as well as viruses from various other families. However, Vero cells generally express high levels of multi-drug resistance 1 (MDR1) or Pgp protein, the efflux transporter of foreign substances including many antiviral compounds, affecting the antiviral activity as well as interpretation of data. To address this, a Pgp gene knockout VeroE6 cell line (VeroE6-Pgp-KO) was generated using CRISPR-CAS9 technology. These cells no longer expressed the Pgp protein as indicated by flow cytometry analysis following staining with a Pgp-specific monoclonal antibody. They also showed significantly reduced efflux transporter activity in the calcein acetoxymethyl ester (calcein AM) assay. The VeroE6-Pgp-KO cells and the parental VeroE6 cells were each infected with SARS-CoV-2 to test antiviral activities of remdesivir and nirmatrelvir, two known Pgp substrates, in the presence or absence of a Pgp inhibitor. The compounds showed antiviral activities in VeroE6-Pgp-KO cells similar to that observed in the presence of the Pgp inhibitor. Thus, the newly established VeroE6-Pgp-KO cell line adds a new in vitro virus infection system for SARS-CoV-2 and possibly other viruses to test antiviral therapies without a need to control the Pgp activity. Removal of the Pgp inhibitor for antiviral assays will lead to less data variation and prevent failed assays.
- Published
- 2022
34. An oral SARS-CoV-2 M
- Author
-
Dafydd R, Owen, Charlotte M N, Allerton, Annaliesa S, Anderson, Lisa, Aschenbrenner, Melissa, Avery, Simon, Berritt, Britton, Boras, Rhonda D, Cardin, Anthony, Carlo, Karen J, Coffman, Alyssa, Dantonio, Li, Di, Heather, Eng, RoseAnn, Ferre, Ketan S, Gajiwala, Scott A, Gibson, Samantha E, Greasley, Brett L, Hurst, Eugene P, Kadar, Amit S, Kalgutkar, Jack C, Lee, Jisun, Lee, Wei, Liu, Stephen W, Mason, Stephen, Noell, Jonathan J, Novak, R Scott, Obach, Kevin, Ogilvie, Nandini C, Patel, Martin, Pettersson, Devendra K, Rai, Matthew R, Reese, Matthew F, Sammons, Jean G, Sathish, Ravi Shankar P, Singh, Claire M, Steppan, Al E, Stewart, Jamison B, Tuttle, Lawrence, Updyke, Patrick R, Verhoest, Liuqing, Wei, Qingyi, Yang, and Yuao, Zhu
- Subjects
Mice, Inbred BALB C ,Ritonavir ,Clinical Trials, Phase I as Topic ,Lactams ,Proline ,Viral Protease Inhibitors ,SARS-CoV-2 ,Administration, Oral ,COVID-19 ,Microbial Sensitivity Tests ,Virus Replication ,COVID-19 Drug Treatment ,Coronavirus ,Disease Models, Animal ,Mice ,Leucine ,Nitriles ,Animals ,Humans ,Drug Therapy, Combination ,Randomized Controlled Trials as Topic - Abstract
The worldwide outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic. Alongside vaccines, antiviral therapeutics are an important part of the healthcare response to countering the ongoing threat presented by COVID-19. Here, we report the discovery and characterization of PF-07321332, an orally bioavailable SARS-CoV-2 main protease inhibitor with in vitro pan-human coronavirus antiviral activity and excellent off-target selectivity and in vivo safety profiles. PF-07321332 has demonstrated oral activity in a mouse-adapted SARS-CoV-2 model and has achieved oral plasma concentrations exceeding the in vitro antiviral cell potency in a phase 1 clinical trial in healthy human participants.
- Published
- 2021
35. Maternal immunization with Group B Streptococcus six-valent polysaccharide conjugate vaccine supported by lack of toxicity in rat and rabbit fertility and developmental toxicity studies
- Author
-
Natasha R, Catlin, Gregg D, Cappon, Scott, Engel, Cynthia, Rohde, William S, Nowland, Sandra, Buitrago, Ingrid, Scully, Annaliesa S, Anderson, and Christopher J, Bowman
- Subjects
Rats, Sprague-Dawley ,Fertility ,Vaccines, Conjugate ,Polysaccharides ,Pregnancy ,Animals ,Humans ,Female ,Immunization ,Rabbits ,Rats ,Streptococcus agalactiae - Abstract
A maternal Group B Streptococcus (GBS) six-valent polysaccharide conjugate vaccine (GBS6) is being developed to protect neonates and infants up to 3 months of age through passive transfer of antibodies from the mother to the infant. Fertility and developmental toxicity studies were conducted in female Sprague Dawley rats and New Zealand White rabbits with GBS6 (20 μg capsular polysaccharide/serotype formulated with or without AlPO
- Published
- 2021
36. An Oral SARS-CoV-2 Mpro Inhibitor Clinical Candidate for the Treatment of COVID-19
- Author
-
Alyssa Dantonio, Heather Eng, Nandini Chaturbhai Patel, Anthony Carlo, Liuqing Wei, Annaliesa S. Anderson, Ravi Shankar P. Singh, Lawrence W. Updyke, Amit S. Kalgutkar, Jamison B. Tuttle, Lisa Aschenbrenner, Simon Berritt, Ketan S. Gajiwala, Jack C. Lee, Devendra K. Rai, Martin Pettersson, Matthew R. Reese, Jisun Lee, Stephen W. Mason, Eugene P. Kadar, RoseAnn Ferre, Melissa Avery, Li Di, Charlotte Moira Norfor Allerton, R. Scott Obach, Scott A. Gibson, Stephen Noell, Patrick Robert Verhoest, Kevin Ogilvie, Jonathan J. Novak, Brett L. Hurst, Dafydd R. Owen, Rhonda D. Cardin, Wei Liu, Yuao Zhu, Karen J. Coffman, Claire M. Steppan, Jean G. Sathish, Samantha Elizabeth Greasley, Matthew F. Sammons, Qingyi Yang, Britton Boras, and Al E. Stewart
- Subjects
Protease ,business.industry ,viruses ,medicine.medical_treatment ,Phases of clinical research ,Pharmacology ,medicine.disease_cause ,In vitro ,Clinical trial ,In vivo ,Pandemic ,Medicine ,Protease inhibitor (pharmacology) ,business ,Coronavirus - Abstract
The worldwide outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become an established global pandemic. Alongside vaccines, antiviral therapeutics are an important part of the healthcare response to counter the ongoing threat presented by COVID-19. Here, we report the discovery and characterization of PF-07321332, an orally bioavailable SARS-CoV-2 main protease inhibitor with in vitro pan-human coronavirus antiviral activity, and excellent off-target selectivity and in vivo safety profiles. PF-07321332 has demonstrated oral activity in a mouse- adapted SARS-CoV-2 model and has achieved oral plasma concentrations exceeding the in vitro antiviral cell potency, in a phase I clinical trial in healthy human participants. ClinicalTrials.gov Identifier: NCT04756531 One-Sentence Summary PF-07321332 is disclosed as a novel, orally active, investigational small-molecule inhibitor of the SARS-CoV-2 main protease, which is being evaluated in clinical trials for the treatment of COVID-19.
- Published
- 2021
37. The impact of human vaccines on bacterial antimicrobial resistance. A review
- Author
-
Kathrin U. Jansen, Raphael Simon, Annaliesa S. Anderson, William C. Gruber, and James Wassil
- Subjects
Viral vaccine ,medicine.drug_class ,business.industry ,Antibiotic resistance ,Viral Vaccine ,Antibiotics ,Human pathogen ,Review ,Multidrug resistance ,medicine.disease_cause ,Virology ,Herd immunity ,Bacterial vaccine ,Multiple drug resistance ,Streptococcus pneumoniae ,medicine ,Environmental Chemistry ,Human vaccination ,business - Abstract
At present, the dramatic rise in antimicrobial resistance (AMR) among important human bacterial pathogens is reaching a state of global crisis threatening a return to the pre-antibiotic era. AMR, already a significant burden on public health and economies, is anticipated to grow even more severe in the coming decades. Several licensed vaccines, targeting both bacterial (Haemophilus influenzae type b, Streptococcus pneumoniae, Salmonella enterica serovar Typhi) and viral (influenza virus, rotavirus) human pathogens, have already proven their anti-AMR benefits by reducing unwarranted antibiotic consumption and antibiotic-resistant bacterial strains and by promoting herd immunity. A number of new investigational vaccines, with a potential to reduce the spread of multidrug-resistant bacterial pathogens, are also in various stages of clinical development. Nevertheless, vaccines as a tool to combat AMR remain underappreciated and unfortunately underutilized. Global mobilization of public health and industry resources is key to maximizing the use of licensed vaccines, and the development of new prophylactic vaccines could have a profound impact on reducing AMR.
- Published
- 2021
38. A Phase 2 Study Evaluating the Safety, Tolerability, and Immunogenicity of Two 3-Dose Regimens of a Clostridium difficile Vaccine in Healthy US Adults Aged 65 to 85 Years
- Author
-
Kathrin U. Jansen, James Peterson, Shon A Remich, Chris Webber, William C. Gruber, Yahong Peng, Charles Knirsch, Annaliesa S. Anderson, Michael W. Pride, and Nicholas Kitchin
- Subjects
Male ,0301 basic medicine ,Microbiology (medical) ,medicine.medical_specialty ,Dose ,030106 microbiology ,nosocomial diarrhea ,Placebo ,Drug Administration Schedule ,03 medical and health sciences ,Immunogenicity, Vaccine ,0302 clinical medicine ,Clostridium difficile infection ,Internal medicine ,adults ,medicine ,Humans ,030212 general & internal medicine ,Adverse effect ,Articles and Commentaries ,Aged ,Aged, 80 and over ,Clostridioides difficile ,business.industry ,Immunogenicity ,Vaccination ,Clostridium difficile ,Antibodies, Bacterial ,Healthy Volunteers ,United States ,Clinical trial ,Regimen ,Infectious Diseases ,Tolerability ,Bacterial Vaccines ,Clostridium Infections ,Female ,toxoid vaccine ,business - Abstract
Background Clostridium difficile causes toxin-mediated nosocomial diarrhea and community-acquired infections; no preventive vaccine is licensed. In this phase 2 study, we explored safety, tolerability, and immunogenicity in older US adults of an investigational bivalent C. difficile vaccine that contains equal dosages of genetically and chemically detoxified toxins A and B. Methods Conducted from July 2015 through March 2017, 855 healthy adults aged 65–85 years from 15 US centers were randomized 3:3:1 to receive vaccine (100 or 200 μg) or placebo at 0, 1, and 6 months (month regimen) or 1, 8, and 30 days (day regimen). Serum toxin A– and B–specific neutralizing antibodies were measured. Participant-reported local reactions (LRs) and systemic events (SEs), adverse events (AEs), serious AEs, newly diagnosed chronic medical conditions, and immediate AEs were recorded. Results The 200-μg dose level elicited higher immune responses than the 100-µg dose level across regimens. Compared with the day regimen, the month regimen induced stronger and more persistent immune responses that remained elevated 12 months after dose 3. Responses peaked at month 7 (month regimen) and day 37 (day regimen). LRs (primarily injection site pain) were more frequent in vaccine recipients than controls; SE frequency was similar across groups. More related AEs were reported in the day regimen group than the month regimen group. Conclusions The C. difficile vaccine was safe, well tolerated, and immunogenic in healthy US adults aged 65–85 years. Immune responses were particularly robust in the 200-μg month regimen group. These results support continued vaccine development. Clinical Trials Registration NCT02561195., Prevention of Clostridium difficile infection is a significant unmet medical need. Here, a 3-dose series of an investigational C. difficile vaccine provided robust immune responses in older US adults. The vaccine was generally well tolerated, supporting its continued clinical development.
- Published
- 2019
39. A Novel Hexavalent Capsular Polysaccharide Conjugate Vaccine (GBS6) for the Prevention of Neonatal Group B Streptococcal Infections by Maternal Immunization
- Author
-
Srinivas Kodali, Jin-Hwan Kim, Jianxin Gu, Ed T. Buurman, Yongdong Liu, Handke Luke David, Suddham Singh, Annaliesa S. Anderson, Danka Pavliakova, Christine Singer, Jason Arnold Lotvin, Mininni Terri L, Yekaterina Timofeyeva, A. Krishna Prasad, Kathrin U. Jansen, Ingrid L. Scully, Robert G. K. Donald, and Soraya Moghazeh
- Subjects
group B streptococcus ,0301 basic medicine ,Group B Streptococcal Infection ,CRM197 ,Serogroup ,Active immunization ,medicine.disease_cause ,Immunoglobulin G ,Streptococcus agalactiae ,maternal vaccine ,Mice ,Major Articles and Brief Reports ,03 medical and health sciences ,conjugate ,0302 clinical medicine ,GBS6 ,Conjugate vaccine ,Streptococcal Infections ,medicine ,Animals ,Immunology and Allergy ,030212 general & internal medicine ,Vaccines, Conjugate ,Bacteria ,biology ,business.industry ,Polysaccharides, Bacterial ,Streptococcal Vaccines ,Vaccination ,Streptococcus ,Antibodies, Bacterial ,Macaca mulatta ,capsular polysaccharide ,030104 developmental biology ,Infectious Diseases ,Animals, Newborn ,Immunization ,Immunology ,biology.protein ,Female ,Antibody ,business ,Immunity, Maternally-Acquired - Abstract
Background Group B streptococcus (GBS) causes serious diseases in newborn infants, often resulting in lifelong neurologic impairments or death. Prophylactic vaccination of pregnant women prior to delivery could provide comprehensive protection, as early onset and late-onset disease and maternal complications potentially could be addressed. Methods Capsular polysaccharide conjugate vaccine GBS6 was designed using surveillance data yielded by whole-genome sequencing of a global collection of recently recovered GBS isolates responsible for invasive neonatal GBS disease. Capsular polysaccharides were isolated, oxidized using sodium periodate, and conjugated to CRM197 by reductive amination in dimethyl sulfoxide. Immune responses in mice and rhesus macaques were measured in a multiplex Luminex immunoglobulin G (IgG) assay and opsonophagocytic activity assays. Results The optimized conjugates were immunogenic, alone and in combination, in mice and rhesus macaques, inducing IgG antibodies that mediated opsonophagocytic killing. Active immunization of murine dams with GBS6 prior to mating resulted in serotype-specific protection of pups from a lethal challenge with GBS. Protection following passive administration of serotype-specific IgG monoclonal antibodies to dams demonstrated conclusively that anticapsular polysaccharide IgG alone is sufficient for protection. Conclusions The findings support the ongoing clinical evaluation of maternal GBS6 vaccination as a potential alternative method to prevent GBS disease in infants., Six-valent capsular polysaccharide conjugate vaccine GBS6 for the prevention of neonatal Group B Streptococcal (GBS) infections was evaluated in preclinical models. Vaccination with GBS6 or administration of serotype-specific IgG to pregnant dams protected pups against lethal challenges with GBS.
- Published
- 2019
40. A lightspeed approach to pandemic drug development
- Author
-
Annaliesa S, Anderson
- Subjects
Drug Development ,Equipment Design ,General Medicine ,Pandemics ,General Biochemistry, Genetics and Molecular Biology - Published
- 2022
41. A phase 3 study to assess the immunogenicity, safety, and tolerability of MenB-FHbp administered as a 2-dose schedule in adolescents and young adults
- Author
-
Daniel Drazan, Hanna Czajka, Jason D. Maguire, Jean-Louis Pregaldien, Roger Maansson, Robert O'Neill, Annaliesa S Anderson, Paul Balmer, Johannes Beeslaar, and John L Perez
- Subjects
General Veterinary ,General Immunology and Microbiology ,Adolescent ,Vaccination ,Public Health, Environmental and Occupational Health ,Meningococcal Vaccines ,Neisseria meningitidis ,Neisseria meningitidis, Serogroup B ,Serogroup ,Antibodies, Bacterial ,Meningococcal Infections ,Young Adult ,Infectious Diseases ,Molecular Medicine ,Humans - Abstract
The MenB-FHbp vaccine is licensed to prevent meningococcal serogroup B disease on either a 2-dose (0, 6 months) or 3-dose (0, 1-2, 6 months) series. This phase 3 study further assessed the immunogenicity and safety of the 2-dose MenB-FHbp schedule.Subjects 10-25 years of age received MenB-FHbp (months 0, 6) and the quadrivalent meningococcal conjugate vaccine MenACWY-CRM (month 0). Primary immunogenicity endpoints included percentages of subjects achieving ≥ 4-fold increases from baseline in serum bactericidal antibody using human complement (hSBA) titers for 4 diverse, vaccine-heterologous primary serogroup B test strains and titers ≥ lower limit of quantitation (LLOQ; 1:8 or 1:16) for all 4 primary strains combined (composite response) after dose 2; a titer ≥ 1:4 is the accepted correlate of protection. Percentages of participants with hSBA titers ≥ LLOQ for 10 additional vaccine-heterologous strains were also assessed; positive predictive values of primary strain responses for secondary strain responses were determined. Safety was assessed.Overall, 1057 subjects received dose 1 and 946 received dose 2 of MenB-FHbp. Percentages of participants achieving ≥ 4-fold increases in hSBA titers against each primary strain after dose 2 ranged from 67.4% to 95.0% and the composite response was 74.3%. Primary strain responses were highly predictive of secondary strain responses. Most reactogenicity events were mild-to-moderate in severity and did not lead to withdrawal from the study. Adverse events (AEs) considered by the investigator to be related to vaccination occurred in 4.2% (44/1057) of subjects, and there were no serious AEs or newly diagnosed chronic medical conditions considered related to vaccination.MenB-FHbp administered at 0, 6 months was well tolerated and induced protective bactericidal antibody responses against diverse serogroup B strains. Findings provide further support for the continued use of MenB-FHbp on a 2-dose schedule in this population.
- Published
- 2021
42. A Comparative Analysis of SARS-CoV-2 Antivirals Characterizes 3CL pro Inhibitor PF-00835231 as a Potential New Treatment for COVID-19
- Author
-
Joseph C. Devlin, Ellie Ivanova, Joseph John Binder, Rachel A Prescott, Annaliesa S. Anderson, Claire M. Steppan, Ana M. Valero-Jimenez, Alberto Herrera, Austin Schinlever, Meike Dittmann, Kelly V. Ruggles, Paige Loose, Adil Mohamed, Ludovic Desvignes, Sergei B. Koralov, Rebecca E. O’Connor, and Maren de Vries
- Subjects
0303 health sciences ,Protease ,biology ,medicine.drug_class ,viruses ,medicine.medical_treatment ,Immunology ,Pharmacology ,Microbiology ,In vitro ,03 medical and health sciences ,0302 clinical medicine ,Viral life cycle ,Virology ,Insect Science ,medicine ,Vero cell ,biology.protein ,Potency ,Respiratory epithelium ,030212 general & internal medicine ,Antiviral drug ,Polymerase ,030304 developmental biology - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of Coronavirus Disease 2019 (COVID-19). There is a dire need for novel effective antivirals to treat COVID-19, as the only approved direct-acting antiviral to date is remdesivir, targeting the viral polymerase complex. A potential alternate target in the viral life cycle is the main SARS-CoV-2 protease 3CLpro (Mpro). The drug candidate PF-00835231 is the active compound of the first anti-3CLpro regimen in clinical trials. Here, we perform a comparative analysis of PF-00835231, the pre-clinical 3CLpro inhibitor GC-376, and the polymerase inhibitor remdesivir, in alveolar basal epithelial cells modified to express ACE2 (A549+ACE2 cells). We find PF-00835231 with at least similar or higher potency than remdesivir or GC-376. A time-of-drug-addition approach delineates the timing of early SARS-CoV-2 life cycle steps in A549+ACE2 cells and validates PF-00835231's early time of action. In a model of the human polarized airway epithelium, both PF-00835231 and remdesivir potently inhibit SARS-CoV-2 at low micromolar concentrations. Finally, we show that the efflux transporter P-glycoprotein, which was previously suggested to diminish PF-00835231's efficacy based on experiments in monkey kidney Vero E6 cells, does not negatively impact PF-00835231 efficacy in either A549+ACE2 cells or human polarized airway epithelial cultures. Thus, our study provides in vitro evidence for the potential of PF-00835231 as an effective SARS-CoV-2 antiviral and addresses concerns that emerged based on prior studies in non-human in vitro models.Importance:The arsenal of SARS-CoV-2 specific antiviral drugs is extremely limited. Only one direct-acting antiviral drug is currently approved, the viral polymerase inhibitor remdesivir, and it has limited efficacy. Thus, there is a substantial need to develop additional antiviral compounds with minimal side effects and alternate viral targets. One such alternate target is its main protease, 3CLpro (Mpro), an essential component of the SARS-CoV-2 life cycle processing the viral polyprotein into the components of the viral polymerase complex. In this study, we characterize a novel antiviral drug, PF-00835231, which is the active component of the first-in-class 3CLpro-targeting regimen in clinical trials. Using 3D in vitro models of the human airway epithelium, we demonstrate the antiviral potential of PF-00835231 for inhibition of SARS-CoV-2.
- Published
- 2021
43. A comparative analysis of SARS-CoV-2 antivirals characterizes 3CL
- Author
-
Maren, de Vries, Adil S, Mohamed, Rachel A, Prescott, Ana M, Valero-Jimenez, Ludovic, Desvignes, Rebecca, O'Connor, Claire, Steppan, Joseph C, Devlin, Ellie, Ivanova, Alberto, Herrera, Austin, Schinlever, Paige, Loose, Kelly, Ruggles, Sergei B, Koralov, Annaliesa S, Anderson, Joseph, Binder, and Meike, Dittmann
- Subjects
viruses ,fungi ,virus diseases ,skin and connective tissue diseases ,Article ,respiratory tract diseases - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of Coronavirus Disease 2019 (COVID-19), a pandemic that has claimed over 700,000 human lives. The only SARS-CoV-2 antiviral, for emergency use, is remdesivir, targeting the viral polymerase complex. PF-00835231 is a pre-clinical lead compound with an alternate target, the main SARS-CoV-2 protease 3CLpro (Mpro). Here, we perform a comparative analysis of PF-00835231 and remdesivir in A549+ACE2 cells, using isolates of two major SARS-CoV-2 clades. PF-00835231 is antiviral for both clades, and, in this assay, statistically more potent than remdesivir. A time-of-drug-addition approach delineates the timing of early SARS-CoV-2 life cycle steps and validates PF-00835231’s time of action. Both PF-00835231 and remdesivir potently inhibit SARS-CoV-2 in human polarized airway epithelial cultures. Thus, our study provides in vitro evidence for the potential of PF-00835231 as an effective antiviral for SARS-CoV-2, addresses concerns from non-human in vitro models, and supports further studies with this compound
- Published
- 2021
44. Preclinical characterization of an intravenous coronavirus 3CL protease inhibitor for the potential treatment of COVID19
- Author
-
Heather Eng, Annaliesa S. Anderson, Norimitsu Shirai, Brandon J. Anson, James Logue, Stuart Weston, Marisa McGrath, Martyn D. Ticehurst, Rebecca E. O’Connor, Michelle Rossulek, Martin Pettersson, Matthew N O' Brien, Jean G. Sathish, Matthew B. Frieman, Emi Kimoto, Jun Wang, R. Scott Obach, Emily I. Chen, Robert Haupt, Yuao Zhu, Thomas F. Rogers, Andrew D. Mesecar, Suman Luthra, Adolfo García-Sastre, Dafydd R. Owen, Rhys M. Jones, Eugene P. Kadar, Chunlong Ma, Rob Kania, Lisa Aschenbrenner, Arnab K. Chatterjee, Charlotte Moira Norfor Allerton, Joseph John Binder, Kevin Ogilvie, Holly L. Hammond, Nathan Beutler, Claire M. Steppan, Jennifer Hammond, Stephen Noell, Romel Rosales, Robert M. Hoffman, Lillis Jonathan Richard, Matthew R. Reese, Stephen W. Mason, Dan Arenson, Malina A. Bakowski, Lawrence W. Updyke, Lorraine F. Lanyon, Kris M. White, Emma K. Lendy, Melanie G. Kirkpatrick, and Britton Boras
- Subjects
Indoles ,Science ,medicine.medical_treatment ,viruses ,General Physics and Astronomy ,Pharmacology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,Mice ,In vivo ,Coronavirus 229E, Human ,Leucine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Protease inhibitor (pharmacology) ,skin and connective tissue diseases ,Infusions, Intravenous ,Vero Cells ,Coronavirus 3C Proteases ,Coronavirus ,ADME ,Multidisciplinary ,Protease ,Alanine ,Chemistry ,SARS-CoV-2 ,fungi ,COVID-19 ,Drug Synergism ,General Chemistry ,Prodrug ,In vitro ,Adenosine Monophosphate ,Pyrrolidinones ,respiratory tract diseases ,COVID-19 Drug Treatment ,body regions ,Disease Models, Animal ,Coronavirus Protease Inhibitors ,Severe acute respiratory syndrome-related coronavirus ,Drug Design ,Drug Therapy, Combination ,HeLa Cells - Abstract
COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. PF-00835231, a 3CL protease inhibitor, has exhibited potent in vitro antiviral activity against SARS-CoV-2 as a single agent. Here we report, the design and characterization of a phosphate prodrug PF-07304814 to enable the delivery and projected sustained systemic exposure in human of PF-00835231 to inhibit coronavirus family 3CL protease activity with selectivity over human host protease targets. Furthermore, we show that PF-00835231 has additive/synergistic activity in combination with remdesivir. We present the ADME, safety, in vitro, and in vivo antiviral activity data that supports the clinical evaluation of PF-07304814 as a potential COVID-19 treatment.
- Published
- 2021
45. Performance of a Four-Antigen Staphylococcus aureus Vaccine in Preclinical Models of Invasive S. aureus Disease
- Author
-
Yekaterina Timofeyeva, Kathrin U. Jansen, Peimin Lu, Arthur Illenberger, Paul A. Liberator, Annaliesa S. Anderson, and Ingrid L. Scully
- Subjects
0301 basic medicine ,Microbiology (medical) ,conjugated polysaccharide ,Staphylococcus aureus ,medicine.disease_cause ,Microbiology ,Sepsis ,sepsis ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Virology ,medicine ,Endocarditis ,030212 general & internal medicine ,lcsh:QH301-705.5 ,ClfA ,business.industry ,medicine.disease ,protection ,Clumping factor A ,animal models ,MntC ,Vaccination ,surgery-associated infection ,030104 developmental biology ,lcsh:Biology (General) ,Bacteremia ,invasive disease ,SA4Ag vaccine ,business ,Progressive disease - Abstract
A Staphylococcus aureus four-antigen vaccine (SA4Ag) was designed for the prevention of invasive disease in surgical patients. The vaccine is composed of capsular polysaccharide type 5 and type 8 CRM197 conjugates, a clumping factor A mutant (Y338A-ClfA) and manganese transporter subunit C (MntC). S. aureus pathogenicity is characterized by an ability to rapidly adapt to the host environment during infection, which can progress from a local infection to sepsis and invasion of distant organs. To test the protective capacity of the SA4Ag vaccine against progressive disease stages of an invasive S. aureus infection, a deep tissue infection mouse model, a bacteremia mouse model, a pyelonephritis model, and a rat model of infectious endocarditis were utilized. SA4Ag vaccination significantly reduced the bacterial burden in deep tissue infection, in bacteremia, and in the pyelonephritis model. Complete prevention of infection was demonstrated in a clinically relevant endocarditis model. Unfortunately, these positive preclinical findings with SA4Ag did not prove the clinical utility of SA4Ag in the prevention of surgery-associated invasive S. aureus infection.
- Published
- 2021
- Full Text
- View/download PDF
46. The Role of Vaccines in Combating Antimicrobial Resistance
- Author
-
Kathrin U. Jansen, Raphael Simon, Annaliesa S. Anderson, James Wassil, and William C. Gruber
- Subjects
medicine.medical_specialty ,medicine.drug_class ,business.industry ,Public health ,Whole cell vaccine ,Antibiotics ,Antimicrobial ,EXTENSIVE DRUG RESISTANCE ,Herd immunity ,Antibiotic resistance ,Agriculture ,Development economics ,medicine ,Business - Abstract
The introduction of new classes of antibiotics, and the high use of antimicrobials in healthcare, agriculture, and the food industry, have all contributed to accelerate the development of antimicrobial resistance (AMR) in bacterial species and dissemination of antibiotic resistant bacterial strains worldwide. At present, the dramatic rise in AMR among important human bacterial pathogens is reaching a state of global crisis threatening a return to the pre-antibiotic era. AMR, already a significant burden on public health and economies, is anticipated to grow even more severe in the coming decades.
- Published
- 2021
47. Report of the National Institutes of Health SARS-CoV-2 Antiviral Therapeutics Summit
- Author
-
Kumar Singh Saikatendu, Michael J. Sofia, Matthew D. Disney, David Baker, Jennifer O. Nwankwo, Marla Weetall, Annaliesa S. Anderson, Christopher P. Austin, Mindy I. Davis, Matthias Götte, Emmie de Wit, Andrew D. Mesecar, Matthew D. Hall, Richard J. Whitley, Stephanie Moore, James M. Anderson, Kara Carter, George R. Painter, Anthony J. Conley, Charlotte A. Lanteri, Sandra K. Weller, Jay Bradner, Celia A. Schiffer, Tomas Cihlar, Abigail Grossman, Timothy P. Sheahan, Kizzmekia S. Corbett, Stephanie L. Ford-Scheimer, Kyle R. Brimacombe, Lillian Chiang, Elizabeth A. Campbell, Daria J. Hazuda, Mark R. Denison, Frederick G. Hayden, Sara Cherry, Pei Yong Shi, Courtney V. Fletcher, Hilary D. Marston, Jules O'Rear, Hugh D. C. Smyth, Francis S. Collins, and Anthony S. Fauci
- Subjects
0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine.disease_cause ,NIH Virtual SARS-CoV-2 Supplement ,03 medical and health sciences ,0302 clinical medicine ,Research community ,preclinical ,Immunology and Allergy ,Medicine ,State of the science ,Coronavirus ,Medical education ,geography ,Summit ,geography.geographical_feature_category ,SARS-CoV-2 ,business.industry ,Research needs ,antiviral therapeutics ,viral replication machinery ,emerging modalities ,AcademicSubjects/MED00290 ,030104 developmental biology ,Infectious Diseases ,Drug development ,proteases ,business ,030217 neurology & neurosurgery - Abstract
The NIH Virtual SARS-CoV-2 Antiviral Summit, held on 6 November 2020, was organized to provide an overview on the status and challenges in developing antiviral therapeutics for coronavirus disease 2019 (COVID-19), including combinations of antivirals. Scientific experts from the public and private sectors convened virtually during a live videocast to discuss severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) targets for drug discovery as well as the preclinical tools needed to develop and evaluate effective small-molecule antivirals. The goals of the Summit were to review the current state of the science, identify unmet research needs, share insights and lessons learned from treating other infectious diseases, identify opportunities for public-private partnerships, and assist the research community in designing and developing antiviral therapeutics. This report includes an overview of therapeutic approaches, individual panel summaries, and a summary of the discussions and perspectives on the challenges ahead for antiviral development.
- Published
- 2021
- Full Text
- View/download PDF
48. Performance of a Four-Antigen
- Author
-
Ingrid L, Scully, Yekaterina, Timofeyeva, Arthur, Illenberger, Peimin, Lu, Paul A, Liberator, Kathrin U, Jansen, and Annaliesa S, Anderson
- Subjects
surgery-associated infection ,sepsis ,Staphylococcus aureus ,conjugated polysaccharide ,invasive disease ,SA4Ag vaccine ,protection ,Article ,animal models ,ClfA ,MntC - Abstract
A Staphylococcus aureus four-antigen vaccine (SA4Ag) was designed for the prevention of invasive disease in surgical patients. The vaccine is composed of capsular polysaccharide type 5 and type 8 CRM197 conjugates, a clumping factor A mutant (Y338A-ClfA) and manganese transporter subunit C (MntC). S. aureus pathogenicity is characterized by an ability to rapidly adapt to the host environment during infection, which can progress from a local infection to sepsis and invasion of distant organs. To test the protective capacity of the SA4Ag vaccine against progressive disease stages of an invasive S. aureus infection, a deep tissue infection mouse model, a bacteremia mouse model, a pyelonephritis model, and a rat model of infectious endocarditis were utilized. SA4Ag vaccination significantly reduced the bacterial burden in deep tissue infection, in bacteremia, and in the pyelonephritis model. Complete prevention of infection was demonstrated in a clinically relevant endocarditis model. Unfortunately, these positive preclinical findings with SA4Ag did not prove the clinical utility of SA4Ag in the prevention of surgery-associated invasive S. aureus infection.
- Published
- 2020
49. Commentary: Variant Signal Peptides of Vaccine Antigen, FHbp, Impair Processing Affecting Surface Localization and Antibody-Mediated Killing in Most Meningococcal Isolates
- Author
-
Paul Balmer, Annaliesa S. Anderson, Robert G. K. Donald, Jamie Findlow, and Paul A. Liberator
- Subjects
Microbiology (medical) ,Signal peptide ,meningoccocus ,Neisseria meningitidis ,lipoprotein ,lcsh:QR1-502 ,Biology ,Vaccine antigen ,FHbp ,medicine.disease_cause ,serogroup B ,Lnt ,Microbiology ,Virology ,Trumenba® ,lcsh:Microbiology ,vaccine ,Slam ,medicine ,biology.protein ,signal peptide ,Antibody ,Original Research - Abstract
Meningococcal lipoprotein, Factor H binding protein (FHbp), is the sole antigen of the Trumenba vaccine (Pfizer) and one of four antigens of the Bexsero vaccine (GSK) targeting Neisseria meningitidis serogroup B isolates. Lipidation of FHbp is assumed to occur for all isolates. We show in the majority of a collection of United Kingdom isolates (1742/1895) non-synonymous single nucleotide polymorphisms (SNPs) in the signal peptide (SP) of FHbp. A single SNP, common to all, alters a polar amino acid that abolishes processing: lipidation and SP cleavage. Whilst some of the FHbp precursor is retained in the cytoplasm due to reduced binding to SecA, remarkably some is translocated and further surface-localized by Slam. Thus we show Slam is not lipoprotein-specific. In a panel of isolates tested, the overall reduced surface localization of the precursor FHbp, compared to isolates with an intact SP, corresponded with decreased susceptibility to antibody-mediated killing. Our findings shed new light on the canonical pathway for lipoprotein processing and translocation of important relevance for lipoprotein-based vaccines in development and in particular for Trumenba.
- Published
- 2020
50. Discovery of a Novel Inhibitor of Coronavirus 3CL Protease for the Potential Treatment of COVID-19
- Author
-
Emma K. Lendy, Martyn D. Ticehurst, Robert Steven Kania, Lisa Aschenbrenner, Chuang Ma, Michelle Rossulek, Emily I. Chen, Charlotte Moira Norfor Allerton, Rhys M. Jones, Stephen W. Mason, Kevin Ogilvie, Heather Eng, Dan Arenson, Lorraine F. Lanyon, Abhishek Chatterjee, Rob Haupt, Martin Pettersson, Britton Boras, Eugene P. Kadar, Malina A. Bakowski, Yuao Zhu, Obach Rs, Suman Luthra, Stuart Weston, Joseph John Binder, Lillis, Stephen Noell, Thomas F. Rogers, Dafydd R. Owen, O’Brien Mn, Claire M. Steppan, Lawrence W. Updyke, Jennifer Hammond, Jun Wang, Norimitsu Shirai, Brandon J. Anson, Nathan Beutler, Jean G. Sathish, Melanie G. Kirkpatrick, Annaliesa S. Anderson, James Logue, Matthew B. Frieman, Andrew D. Mesecar, Holly L. Hammond, Robert M. Hoffman, Reese Mr, Marisa McGrath, Rebecca E. O’Connor, and Emi Kimoto
- Subjects
Protease ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,Chemistry ,medicine.medical_treatment ,Prodrug ,medicine.disease_cause ,Virology ,Article ,In vitro ,Virus ,Pharmacodynamics ,Preclinical research ,In vivo ,medicine ,Pharmacokinetics ,ADME ,Coronavirus - Abstract
COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. PF-00835231, a 3CL protease inhibitor, has exhibited potent in vitro antiviral activity against SARS-CoV-2 as a single agent. Here we report, the design and characterization of a phosphate prodrug PF-07304814 to enable the delivery and projected sustained systemic exposure in human of PF-00835231 to inhibit coronavirus family 3CL protease activity with selectivity over human host protease targets. Furthermore, we show that PF-00835231 has additive/synergistic activity in combination with remdesivir. We present the ADME, safety, in vitro, and in vivo antiviral activity data that supports the clinical evaluation of PF-07304814 as a potential COVID-19 treatment., The 3CL protease of SARS-CoV-2 is inhibited by PF-00835231 in vitro. Here, the authors show that the prodrug PF-07304814 has broad spectrum activity, inhibiting SARS-CoV and SARS-CoV-2 in mice and its ADME and safety profile support clinical development.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.