Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Open Forum Infectious Diseases ; 9(Supplement 2):S483-S484, 2022.
Article in English | EMBASE | ID: covidwho-2189785

ABSTRACT

Background. ADI is a fully human IgG1 monoclonal antibody engineered to have an extended half-life with high potency and broad neutralization against SARS-CoV-2 and other SARS-like coronaviruses. The goal of our analysis was to develop a QSP model in which ADI concentrations in upper airway (UA) epithelial lining fluid (ELF) were linked to a viral dynamic model to describe the impact of ADI on SARS-CoV-2 viral load relative to placebo. Methods. The QSP model was fit inNONMEMVersion 7.4 using PK data from a Phase 1 study (N=24, IV and IM) and from Phase 2/3 COVID-19 prevention (EVADE;N=659, IM) and treatment (STAMP;N=189, IM) studies. Saliva and NP samples were collected from STAMP study participants (pts) infected with the delta or omicron variants. The viral dynamic model was based on a published model and was modified to include both active (V) and deactivated (DV) virus (Fig). The viral dynamic model was fit to the NP swab viral load data (2 samples/pt) standardized to time since infection based upon recorded symptom onset. Saliva data (7-8 samples/ pt) was fit sequentially using a biophase compartment given the peak viral load was modestly lower and peaked later than Day 1. Viral dynamic model (A) and simulated median (90% PI) NP viral load reduction in ADI-treated or placebo participants for delta (B) and omicron (C) variants Results. The QSP model provided an excellent fit to serum ADI concentrationtime data after estimation of a transit rate to account for IM absorption, plasma volume, and the ADI-neonatal Fc receptor dissociation rate constant. The linked viral dynamic model captured the NP swab viral load data after estimating differences in within-host replication factor (R0) and viral production rate (p) by variant. Maximal ADI-induced effect (Smax) on stimulating viral clearance (c) was fixed to 0.43 based upon prior modeling. ADI concentration in UA ELF resulting in 50% of Smax (SC50) was estimated to be 0.086 for delta and 1.05 mg/L for omicron. Figure B and C show model-based simulated median (90% PI) viral load reduction in ADI-treated or placebo pts for delta and omicron variants. Conclusion. This QSP model, in conjunction with information on new variants available early in outbreaks (IC50, infectivity (R0), viral production rate [each a model parameter]), allows for rapid dose identification in response to emerging variants.

2.
J Transl Med ; 20(1): 146, 2022 03 29.
Article in English | MEDLINE | ID: covidwho-2098352

ABSTRACT

BACKGROUND: The kallikrein-kinin system is assumed to have a multifunctional role in health and disease, but its in vivo role in humans currently remains unclear owing to the divergence of plasma kinin level data published ranging from the low picomolar to high nanomolar range, even in healthy volunteers. Moreover, existing data are often restricted on reporting levels of single kinins, thus neglecting the distinct effects of active kinins on bradykinin (BK) receptors considering diverse metabolic pathways. A well-characterized and comprehensively evaluated healthy cohort is imperative for a better understanding of the biological variability of kinin profiles to enable reliable differentiation concerning disease-specific kinin profiles. METHODS: To study biological levels and variability of kinin profiles comprehensively, 28 healthy adult volunteers were enrolled. Nasal lavage fluid and plasma were sampled in customized protease inhibitor prespiked tubes using standardized protocols, proven to limit inter-day and interindividual variability significantly. Nine kinins were quantitatively assessed using validated LC-MS/MS platforms: kallidin (KD), Hyp4-KD, KD1-9, BK, Hyp3-BK, BK1-8, BK1-7, BK1-5, and BK2-9. Kinin concentrations in nasal epithelial lining fluid were estimated by correlation using urea. RESULTS: Circulating plasma kinin levels were confirmed in the very low picomolar range with levels below 4.2 pM for BK and even lower levels for the other kinins. Endogenous kinin levels in nasal epithelial lining fluids were substantially higher, including median levels of 80.0 pM for KD and 139.1 pM for BK. Hydroxylated BK levels were higher than mean BK concentrations (Hyp3-BK/BK = 1.6), but hydroxylated KD levels were substantially lower than KD (Hyp4-KD/KD = 0.37). No gender-specific differences on endogenous kinin levels were found. CONCLUSIONS: This well-characterized healthy cohort enables investigation of the potential of kinins as biomarkers and would provide a valid control group to study alterations of kinin profiles in diseases, such as angioedema, sepsis, stroke, Alzheimer's disease, and COVID-19.


Subject(s)
Kinins , Chromatography, Liquid , Humans , Kinins/analysis , Receptors, Bradykinin/metabolism , Tandem Mass Spectrometry
3.
Hong Kong Journal of Paediatrics ; 27(1):67, 2022.
Article in English | EMBASE | ID: covidwho-2003052

ABSTRACT

Vaccines that elicit mucosal immune responses against SARS-CoV-2 could potent ially be of exceptional importance in providing first line defence at the site of viral entry. In order to understand the mucosal immune response profiles of SARS-CoV-2 vaccines, we examined both the mucosal and systemic responses of subjects vaccinated by two different vaccination platforms: mRNA (Comirnaty) and inactivated virus (CoronaVac). Nasal epithelial lining fluid (NELF) and peripheral blood samples were collected in subjects who had received two doses of CoronaVac or Comirnaty. We quantified IgA and IgG specific to SARS-CoV-2 S1 protein, neutralisation antibody by ELISA in NELF and plasma samples. Only Comirnaty induced nasal S1-specific IgA and IgG responses, which were evident as early as on 14±2 days after the first dose. The NELF samples of 72% of subjects became IgA+IgG+, while in 62.5% of subjects the samples were neutralising by 7±2 days after the second dose. In 45% of the subjects their NELF remained neutralising 50 days after the booster. In plasma, 91% and 100% Comirnaty subjects possessed S1-specific IgA+IgG+ on 14±2 days after the first dose and 7±2 days after booster, respectively. The plasma collected on 7±2 days after booster was 100% neutralising. The induction of S1-specific antibody by CoronaVac was IgG dominant, and 70% of the subjects possessed specific IgG by 7±2 days after booster and were all neutralising. This study reveals that Comirnaty is able to induce S1-specific IgA and IgG r esponse with neutralising activity in the nasal mucosa in addition to a consistent systemic response.

4.
Antimicrob Agents Chemother ; 66(7): e0059022, 2022 07 19.
Article in English | MEDLINE | ID: covidwho-1909571

ABSTRACT

Tebipenem pivoxil hydrobromide (TBP-PI-HBr) is an oral carbapenem prodrug being developed for the treatment of serious bacterial infections. The active moiety, tebipenem, has broad-spectrum activity against common Enterobacterales pathogens, including extended-spectrum-ß-lactamase (ESBL)-producing multidrug-resistant strains. This study evaluated the intrapulmonary pharmacokinetics (PK) and epithelial lining fluid (ELF) and alveolar macrophage (AM) concentrations of tebipenem relative to plasma levels in nonsmoking, healthy adult subjects. Thirty subjects received oral TBP-PI-HBr at 600 mg every 8 h for five doses. Serial blood samples were collected following the last dose. Each subject underwent one standardized bronchoscopy with bronchoalveolar lavage (BAL) 1, 2, 4, 6, or 8 h after the fifth dose of TBP-PI-HBr. The tebipenem area under the concentration-time curve for the 8-h dosing interval (AUC0-8) values in plasma, ELF, and AMs were calculated using the mean concentration at each BAL sampling time. Ratios of AUC0-8 values for total ELF and AMs to those for unbound plasma were determined, using a plasma protein binding value of 42%. Mean values ± standard deviations (SD) of tebipenem maximum (Cmax) and minimum (Cmin) total plasma concentrations were 11.37 ± 3.87 mg/L and 0.043 ± 0.039 mg/L, respectively. Peak tebipenem concentrations in plasma, ELF, and AMs occurred at 1 h and then decreased over 8 h. Ratios of tebipenem AUC0-8 values for ELF and AMs to those for unbound plasma were 0.191 and 0.047, respectively. Four (13.3%) subjects experienced adverse events (diarrhea, fatigue, papule, and coronavirus disease 2019 [COVID-19]); all resolved, and none were severe or serious. Tebipenem is distributed into the lungs of healthy adults, which supports the further evaluation of TBP-PI-HBr for the treatment of lower respiratory tract bacterial infections caused by susceptible pathogens. (This study has been registered at ClinicalTrials.gov under identifier NCT04710407.).


Subject(s)
Anti-Bacterial Agents , COVID-19 , Administration, Oral , Adult , Anti-Bacterial Agents/pharmacokinetics , Bronchoalveolar Lavage Fluid , Carbapenems/metabolism , Humans , Lung/metabolism , Monobactams/metabolism
5.
Open Forum Infectious Diseases ; 8(SUPPL 1):S634-S635, 2021.
Article in English | EMBASE | ID: covidwho-1746329

ABSTRACT

Background. ADG20 is a fully human IgG1 monoclonal antibody engineered to have potent and broad neutralization against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and additional SARS-like CoVs with pandemic potential and an extended half-life. A QSP/PBPK model was constructed using ADG20-specific physiochemical properties and published non-human primate (NHP) and human PK data for other antibodies;it was used to a priori predict and confirm NHP and human PK. Methods. An existing QSP/PBPK model was modified to include 3 distinct lung sub-compartments: upper airway, lower airway, and alveolar tissue (Figure A). Each sub-compartment (Figure B) contained an epithelial lining fluid (ELF) space (Figure B). The model was fit separately to digitized NHP and human serum PK data for 7 extended half-life antibodies to estimate the apparent neonatal Fc receptor (FcRn) binding affinity (KD,FcRn) and bioavailability by drug. Nasopharyngeal swab (upper airway) and lung (lower airway) ELF PK data from 4 additional antibodies were used to optimize a single rate constant for transcytosis in lung. Patches of positive charge was a covariate on the rate of pinocytosis of antibody entry and exit from the endosomal space (Figure B). Observed NHP (ADG20 10 mg/kg IM) and human (ADG20 300 mg IM) PK data collected over the initial 21 days post dose were compared with model forecasts from a 1000-iteration simulation. Results. The distribution of fitted NHP KD,FcRn provided accurate predictions of NHP serum PK data (Figure C). NHP ADG20 KD,FcRn was optimized to be 35.7 nM and human ADG20 KD,FcRn (9.55 nM) was derived using a mean NHP:human KD,FcRn ratio of 3.74 across antibodies. Model-based simulated human serum PK data using inter-subject variability from NHP and actual weight distribution from an ongoing Phase 1 study aligned with initial 21-day data (Figure D). Using an adult CDC weight distribution (45-150 kg), the simulated median exceeded 74 days. Conclusion. The QSP/PBPK model a priori predicted NHP and human ADG20 PK. This innovative QSP-based modeling and simulation approach enabled the evaluation of candidate dose regimens prior to the availability of PK data, supporting the rapid advancement of the ADG20 clinical program during the COVID-19 pandemic.

6.
Open Forum Infectious Diseases ; 8(SUPPL 1):S635, 2021.
Article in English | EMBASE | ID: covidwho-1746328

ABSTRACT

Background. ADG20 is a fully human IgG1 monoclonal antibody engineered to have potent and broad neutralization against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other SARS-like CoVs with pandemic potential and an extended half-life. ADG20 is administered intramuscularly (IM). A QSP/PBPK model was constructed to support dose selection for a Phase 2/3 trial of ambulatory patients with mild to moderate COVID-19 (STAMP: NCT04805671). Methods. A QSP/PBPK model was used to simulate receptor occupancy (RO) and drug exposure in the upper airway (nasopharyngeal/oropharyngeal epithelial lining fluid [ELF] compartment). RO was linked to an existing viral dynamic model to enable the prediction of the natural time course of viral load and the effect of ADG20 on viral clearance and infectivity rate. RO was calculated using: 1) in vitro ADG20-SARS-CoV-2 binding kinetics (association rate constant (kon) of 1.52E+06 M-1•s1 and dissociation rate constant (koff) of 2.81E-04 s-1 from a Biacore assay;2) time course of ADG20 concentrations in ELF;and 3) time course of viral load following ADG20 administration. Molar SARS-CoV-2 viral binding site capacity was calculated assuming 40 spike proteins per virion, 3 binding sites per spike, and an initial viral load of log 107 copies/mL for all patients. The QSP/PBPK model and a 2018 CDC reference body weight distribution (45-150 kg) were used to simulate 1000 concentration-time profiles for a range of candidate ADG20 regimens. ADG20 regimens were evaluated against 2 criteria: 1) ability to attain near complete ( >90%), and durable (28-day) SARS-CoV-2 RO in the ELF;and 2) ability to maintain ELF ADG20 concentrations relative to a concentration (0.5 mg/L) associated with 100% viral growth suppression in an in vitro post-infection assay. Results. A single 300 mg IM ADG20 dose met the dose selection criteria in terms of RO (Figure A) and viral growth suppression (Figure B). Conclusion. These data support the evaluation of an ADG20 300 mg IM dose for the treatment of mild to moderate COVID-19. ADG20 is forecasted to attain near complete ( >90%) SARS-CoV-2 RO in the ELF and maintain ELF ADG20 concentrations above that associated with 100% viral growth suppression in vitro.

7.
Front Immunol ; 12: 744887, 2021.
Article in English | MEDLINE | ID: covidwho-1497079

ABSTRACT

Background: Although the serological antibody responses induced by SARS-CoV-2 vaccines are well characterized, little is known about their ability to elicit mucosal immunity. Objectives: This study aims to examine and compare the mucosal and systemic responses of recipients of two different vaccination platforms: mRNA (Comirnaty) and inactivated virus (CoronaVac). Methods: Serial blood and nasal epithelial lining fluid (NELF) samples were collected from the recipients of either Comirnaty or CoronaVac. The plasma and NELF immunoglobulins A and G (IgA and IgG) specific to SARS-CoV-2 S1 protein (S1) and their neutralization effects were quantified. Results: Comirnaty induced nasal S1-specific immunoglobulin responses, which were evident as early as 14 ± 2 days after the first dose. In 64% of the subjects, the neutralizing effects of NELF persisted for at least 50 days. Moreover, 85% of Comirnaty recipients exhibited S1-specific IgA and IgG responses in plasma by 14 ± 2 days after the first dose. By 7 ± 2 days after the booster, all plasma samples possessed S1-specific IgA and IgG responses and were neutralizing. The induction of S1-specific plasma antibodies by CoronaVac was IgG dominant, and 83% of the subjects possessed S1-specific IgG by 7 ± 2 days after the booster, with neutralizing effects. Conclusion: Comirnaty induces S1-specific IgA and IgG responses with neutralizing activity in the nasal mucosa; a similar response is not seen with CoronaVac. Clinical Implication: The presence of a nasal response with mRNA vaccine may provide additional protection compared with inactivated virus vaccine. However, whether such widespread immunological response may produce inadvertent adverse effects in other tissues warrants further investigation.


Subject(s)
COVID-19 Vaccines/immunology , Immunity, Mucosal , SARS-CoV-2/immunology , Adult , Age Factors , Aged , Antibodies, Neutralizing/analysis , Antibodies, Viral/analysis , COVID-19/immunology , COVID-19/prevention & control , Female , Humans , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Male , Middle Aged , Nasal Mucosa/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology , Young Adult
8.
Infect Disord Drug Targets ; 21(6): e170721188930, 2021.
Article in English | MEDLINE | ID: covidwho-969383

ABSTRACT

INTRODUCTION: In the current scenario of the COVID 19 pandemic, the protective reflexes, namely sneeze and cough, have received great importance. However, it is not in terms of protection but in terms of the spread of infection. The present review tries to bring out the correlation between the physiology of sneeze and cough, taking into consideration the various receptors that initiate the two reflexes, then correlating it with the formation of expelled droplets and the significance of various aspects of droplets that lead to the spread of infection. MATERIAL AND METHODS: For the compilation of the present review, we searched the terms "Physiology of cough", "Physiology of sneeze", "droplets", "aerosols" and "Aerosols in COVID 19". The above-mentioned terms were extensively searched on PubMed, Google Scholar, and google search engine. After reviewing the various available material, the most significant research has been considered for this review. CONCLUSION: Through this review, we conclude that there are various factors responsible for the initiation of sneeze and cough, but in the case of infection, it is mainly the inflammatory reaction that directly stimulates the receptors to produce the reflex outburst air. As the flow of air during expiration is turbulent, it causes damage to the Epithelial Lining Fluid present in the respiratory conduit. In addition, it gets admixed with the saliva in the oropharynx and oral cavity and mucus in the nose to form droplets of various sizes. Large droplets settle close and are responsible for droplet and fomite transmission, but the smaller droplets remain suspended in the air and travel farther distances to cause airborne transmission. The spread of droplet cloud in sneezing may range to 6m or more as compared to cough; hence the concept of 1m to 2m of social distancing does not hold reliable if the patient is sneezing.


Subject(s)
COVID-19 , Respiratory Tract Infections , Air Microbiology , Humans , SARS-CoV-2 , Sneezing
SELECTION OF CITATIONS
SEARCH DETAIL