Kelvin Kai-Wang To’s research while affiliated with Queen Mary Hospital and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (257)


Human Circovirus in Patients with Hepatitis, Hong Kong
  • Article

December 2024

·

12 Reads

Emerging Infectious Diseases

·

Cyril Chik-Yan Yip

·

Jianwen Situ

·

[...]

·



Evolution of JN.1
a A representative phylogenetic tree of SARS-CoV-2 from September 1, 2023, to March 1, 2024. Pre-Omicron variants are labeled as other variants. b Step-wise accumulation of key spike mutations in Omicron XBB.1, EG.5.1, HK.3, BA.2.86, and JN.1 compared to BA.2. Key mutations are color barcoded, and represented as colored bars when present or white bars if absent. Different amino acids are shown in colored bars. The number of key mutations of each variant is summarized at the top. Mutation events are labeled on arrows. R493Q is a reversion mutation. c Summary of the spike mutations of SARS-CoV-2 lineages BA.2*, BA.2.86*, JN.1*, XBB.1*, EG.5.1*, and HK.3* compared to the ancestral SARS-CoV-2 spike protein. Color represents the proportion of each mutation in each variant. d Variant dynamics of Omicron lineages BA.2*, BA.2.86*, JN.1*, XBB.1*, EG.5.1*, and HK.3* worldwide and in different regions in terms of variant frequency. The genome data from September 1, 2023, to March 1, 2024 were analyzed. Note that BA.2* excludes BA.2.86*, XBB.1* does not include EG.5.1*, EG.5.1* does not include HK.3*, and BA.2.86* does not include JN.1* for sequence and mutation analysis.
Immune resistance and antigenicity of BA.2, BA.2.86, JN.1, XBB.1, EG.5.1, and HK.3
a Neutralization curves (left) and heatmap showing the IC50 values (right) of mAbs with Omicron-spike pseudoviruses (n = 3). The n number represents biological repeats. b Neutralization of the indicated Omicron-spike pseudoviruses by sera collected from individuals between days 14 and 28 after XBB reinfection following prior infection with BA.5 virus after receiving two to three doses of inactivated vaccine (n = 16). The horizontal dashed line indicates the detection limit (40-fold). c Antigenic map based on all the breakthrough infection serum neutralization data from Fig. 2b. Virus positions are represented by closed circles, whereas serum positions are shown as open squares. d Neutralization of the indicated Omicron-spike pseudoviruses by sera collected from individuals between days 14 and 28 after JN.1 reinfection following prior infection with BA.5 virus after receiving three doses of inactivated vaccine (n = 16). The horizontal dashed line indicates the detection limit (20-fold). e Antigenic map based on all the breakthrough infection serum neutralization data from Fig. 2d. Virus positions are represented by closed circles, whereas serum positions are shown as open squares. f Sera from infected hamsters were used for neutralization of the indicated Omicron-spike pseudoviruses (n = 6 for BA.2 and JN.1, n = 5 for BA.2.86, XBB.1, EG.5.1, and HK.3). The horizontal dashed line indicates the detection limit (10-fold). g Antigenic map based on the hamster serum neutralization data from Fig. 2f. Virus positions are represented by closed circles, whereas serum positions are shown as open squares. h Principal components analysis (PCA) map based on the hamster serum neutralization data from Fig. 2f. i Sera was obtained from mice immunized with the spike trimer of BA.2, BA.2.86, JN.1, and XBB.1 (n = 8). The horizontal dashed line indicates the detection limit (20-fold). Statistical significance in (b) was determined with the two-tailed Wilcoxon matched-pairs signed rank test. Statistical significance in (f) and (i) was determined with the Kruskal-Wallis test with post hoc Dunn’s multiple comparisons tests. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. NS is not statistically significant. Source data are provided as a Source Data file.
Virological features of BA.2.86/JN.1 and EG.5.1/HK.3 in vitro
a SARS-CoV-2 spikes mediated cell-cell fusion in vitro. 293T cells (effector cells), which were co-transfected with the indicated spike and GFP1-10, were co-cultured with 293T cells co-transfected with hACE2 and GFP11 (target cells) (n = 10). The cells were fixed after 24 hours of incubation. GFP signal intensity was measured by ImageJ. The n number represents biological repeats. b Entry efficiency of pseudoviruses in cell lines. Pseudoviruses entry were quantified by measuring the luciferase signal at 24 h post-transduction (n = 6 for B.1 and n = 7 for other variants). Fold change in the luciferase signal was normalized to the mean luciferase readout of BA.2-spike pseudoviruses. The n number represents biological repeats. c Plaque size of Omicron BA.2, BA.2.86, JN.1, XBB.1, EG.5.1, and HK.3 in VeroE6 cells. VeroE6 cells were challenged with 60 PFU of Omicron BA.2, BA.2.86, JN.1, XBB.1, EG.5.1, and HK.3. The infected cells were fixed with 4% paraformaldehyde at 96 and 120 hpi and stained with crystal violet. Plaque size was measured by Adobe Photoshop (n = 10). The n number represents biological repeats. d Protease usage by pseudoviruses in cell lines. VeroE6-TMPRSS2, Calu3, Caco2, and 293T cells were pre-treated with 10 μM Camostat or E64D for 2 h, followed by transduction with B.1-, BA.2-, BA.2.86-, JN.1-, XBB.1-, EG.5.1-, and HK.3-spike pseudoviruses. At 24 h post-transduction, the level of pseudovirus entry was quantified by measuring the luciferase signal. The fold change was normalized to the mean luciferase readout of cells treated with DMSO for each variant (n = 6). The n number represents biological repeats. Data represent mean ± SEM. Statistical significance in (a–d) was determined with one-way ANOVA with post hoc Tukey’s multiple comparisons test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. NS is not statistically significant. Source data are provided as a Source Data file.
Virological features of BA.2.86/JN.1 and EG.5.1/HK.3 in human nasal epithelial cells
a Replication of BA.2, BA.2.86, JN.1, EG.5.1 and HK.3 in Calu3 cells (n = 6). Cell lysates were quantified for viral subgenomic E gene (sgE) copies. The n number represents biological repeats. b Replication of BA.2, BA.2.86, JN.1, EG.5.1 and HK.3 in hNECs (n = 3). Cell lysates were quantified for viral sgE copies. The n number represents biological repeats. c Pseudovirus entry in hNECs. hNECs were transduced with BA.2.86- (n = 7), JN.1- (n = 7), EG.5.1- (n = 4), and HK.3-spike (n = 4) pseudoviruses. Pseudovirus entries were quantified by measuring the luciferase signal. The n number represents biological repeats. d Preference of protease usage by BA.2.86 and JN.1 in hNECs (n = 3). The supernatant was quantified for viral RdRp gene level with qRT-PCR. The n number represents biological repeats. e Spike cleavage of BA.2.86, JN.1, EG.5.1, and HK.3 in hNECs. A representative image of the spike was shown with β-actin added as a sample processing control. Spike and β-actin were run on different gels and detected on different membranes. The cleavage ratio of spike proteins was quantified by ImageJ. f Quantification of spike cleavage as performed in Fig. 4e (n = 4). The n number represents biological repeats. g Infectivity of BA.2.86, JN.1, EG.5.1, and HK.3 in hNECs. Infected hNECs were fixed at 48 hpi for the visualization of ciliated cell marker beta-tubulin (red) and SARS-CoV-2 nucleocapsid protein (green). The DAPI channel was included in the merged images. Scale bar, 20 μm. h Immunofluorescence signal intensity of SARS-CoV-2 N BA.2.86 (n = 5), JN.1 (n = 9), EG.5.1 (n = 7), and HK.3 (n = 5) in Fig. 4g was quantified with ImageJ. The n number represents biological repeats. Scale bar = 20 µm. Data represent mean ± SEM. Statistical significance in (a–c and f, h) was determined with one-way ANOVA with post hoc Tukey’s multiple comparisons test. Statistical significance in (d) was determined with two-way ANOVA with Sidak’s multiple comparison tests. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. NS is not statistically significant. Source data are provided as a Source Data file.
Cryo-EM structures of BA.2.86/JN.1 spikes in complex with ACE2
a Cryo-EM structure of the BA.2.86 spike in complex with ACE2. Two perpendicular views of BA.2.86 spike-ACE2 are depicted as surface, with ACE2 in cornflower blue and the trimeric spike in pink, medium purple, and purple. b Structure of BA.2.86 RBD-ACE2. c Cryo-EM structure of the JN.1 spike in complex with ACE2. Two perpendicular views of JN.1 spike-ACE2 are depicted as surface, with ACE2 in cornflower blue and the trimeric spike in orange, gold, and pale violet. d Structure of JN.1 RBD-ACE2. e Detailed interactions between the BA.2.86 spikeL455-ACE2 and the local refinement cryo-EM maps. f Detailed interactions between the JN.1 spikeS455-ACE2 and the local refinement cryo-EM maps. g Comparation of the RBD-ACE2 interface region in other recent variants: WT RBD-ACE2(PDB:6LZG), XBB.1 RBD-ACE2(PDB:8IOV), EG.5.1 RBD-ACE(PDB:8XLN).

+1

Lineage-specific pathogenicity, immune evasion, and virological features of SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3
  • Article
  • Full-text available

October 2024

·

81 Reads

SARS-CoV-2 JN.1 with an additional L455S mutation on spike when compared with its parental variant BA.2.86 has outcompeted all earlier variants to become the dominant circulating variant. Recent studies investigated the immune resistance of SARS-CoV-2 JN.1 but additional factors are speculated to contribute to its global dominance, which remain elusive until today. Here, we find that SARS-CoV-2 JN.1 has a higher infectivity than BA.2.86 in differentiated primary human nasal epithelial cells (hNECs). Mechanistically, we demonstrate that the gained infectivity of SARS-CoV-2 JN.1 over BA.2.86 associates with increased entry efficiency conferred by L455S and better spike cleavage in hNECs. Structurally, S455 altered the mode of binding of JN.1 spike protein to ACE2 when compared to BA.2.86 spike at ACE2H34, and modified the internal structure of JN.1 spike protein by increasing the number of hydrogen bonds with neighboring residues. These findings indicate that a single mutation (L455S) enhances virus entry in hNECs and increases immune evasiveness, which contribute to the robust transmissibility of SARS-CoV-2 JN.1. We further evaluate the in vitro and in vivo virological characteristics between SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3, and identify key lineage-specific features of the two Omicron sublineages that contribute to our understanding on Omicron antigenicity, transmissibility, and pathogenicity.

Download



Fig. 1: PRISMA reporting for systematic review. Abbreviations: DNS, de novo amino acid substitutions.
Fig. 3: Frequency of DNS that emerged during chronic infection. a) All viral proteins; b) Different domains of the S protein. The frequency is calculated by the sum of DNS at an amino acid residue of a protein, and divided by the length of the protein. All DNS at a particular amino acid residue for a single patient is counted as one DNS. The dotted line represents the overall frequency of DNS for all proteins (a) or the spike protein (b). # represents proteins that have statistically significantly higher frequency of DNS than the overall frequency of DNS after Bonferroni correction.
The significance of recurrent de novo amino acid substitutions that emerged during chronic SARS-CoV-2 infection: an observational study

August 2024

·

29 Reads

EBioMedicine

Background De novo amino acid substitutions (DNS) frequently emerge among immunocompromised patients with chronic SARS-CoV-2 infection. While previous studies have reported these DNS, their significance has not been systematically studied. Methods We performed a review of DNS that emerged during chronic SARS-CoV-2 infection. We searched PubMed until June 2023 using the keywords “(SARS-CoV-2 or COVID-19) and (mutation or sequencing) and ((prolonged infection) or (chronic infection) or (long term))”. We included patients with chronic SARS-CoV-2 infection who had SARS-CoV-2 sequencing performed for at least 3 time points over at least 60 days. We also included 4 additional SARS-CoV-2 patients with chronic infection of our hospital not reported previously. We determined recurrent DNS that has appeared in multiple patients and determined the significance of these mutations among epidemiologically-significant variants. Findings A total of 34 cases were analyzed, including 30 that were published previously and 4 from our hospital. Twenty two DNS appeared in ≥3 patients, with 14 (64%) belonging to lineage-defining mutations (LDMs) of epidemiologically-significant variants and 10 (45%) emerging among chronically-infected patients before the appearance of the corresponding variant. Notably, nsp9-T35I substitution (Orf1a T4175I) emerged in all three patients with BA.2.2 infection in 2022 before the appearance of Variants of Interest that carry nsp9-T35I as LDM (EG.5 and BA.2.86/JN.1). Structural analysis suggests that nsp9-T35I substitution may affect nsp9-nsp12 interaction, which could be critical for the function of the replication and transcription complex. Interpretation DNS that emerges recurrently in different chronically-infected patients may be used as a marker for potential epidemiologically-significant variants. Funding Theme-Based Research Scheme [T11/709/21-N] of the 10.13039/501100002920Research Grants Council (See acknowledgements for full list).


SARS-CoV-2 variants divergently infectand damage cardiomyocytes in vitro and in vivo

August 2024

·

98 Reads

Cell & Bioscience

Background COVID-19 can cause cardiac complications and the latter are associated with poor prognosis and increased mortality. SARS-CoV-2 variants differ in their infectivity and pathogenicity, but how they affect cardiomyocytes (CMs) is unclear. Methods The effects of SARS-CoV-2 variants were investigated using human induced pluripotent stem cell-derived (hiPSC-) CMs in vitro and Golden Syrian hamsters in vivo. Results Different variants exhibited distinct tropism, mechanism of viral entry and pathology in the heart. Omicron BA.2 most efficiently infected and injured CMs in vitro and in vivo , and induced expression changes consistent with increased cardiac dysfunction, compared to other variants tested. Bioinformatics and upstream regulator analyses identified transcription factors and network predicted to control the unique transcriptome of Omicron BA.2 infected CMs. Increased infectivity of Omicron BA.2 is attributed to its ability to infect via endocytosis, independently of TMPRSS2, which is absent in CMs. Conclusions In this study, we reveal previously unknown differences in how different SARS-CoV-2 variants affect CMs. Omicron BA.2, which is generally thought to cause mild disease, can damage CMs in vitro and in vivo. Our study highlights the need for further investigations to define the pathogenesis of cardiac complications arising from different SARS-CoV-2 variants.


Fig. 3. Comparison of the antibody levels against the spike protein receptor binding domain of ancestral strain and Omicron BA.2 sublineage. Panels A, B, C, and D denote antibody levels of salivary IgA, salivary IgG, serum IgA, and serum IgG, respectively, in non-infected patients, while panels E, F, G, and H represent the same antibody titers in infected patients. Horizontal bar represents the median and interquartile range, and each dot represents the area under curve (AUC) for each study participant. Wilcoxon matched-pairs signed rank test was used in the comparison of antibody level against ancestral strain and Omicron BA.2 strain.
Fig. 4. Correlation between salivary IgA, salivary IgG, serum IgA and serum IgG levels.
Baseline characteristics of patients.
The impact of vaccine type and booster dose on the magnitude and breadth of SARS-CoV-2-specific systemic and mucosal antibodies among COVID-19 vaccine recipients

August 2024

·

15 Reads

Heliyon

The COVID-19 pandemic has had a major impact on global health and economy, which was significantly mitigated by the availability of COVID-19 vaccines. The levels of systemic and mucosal antibodies against SARS-CoV-2 correlated with protection. However, there is limited data on how vaccine type and booster doses affect mucosal antibody response, and how the breadth of mucosal and systemic antibodies compares. In this cross-sectional study, we compared the magnitude and breadth of mucosal and systemic antibodies in 108 individuals who received either the BNT162b2 (Pfizer) or CoronaVac (SinoVac) vaccine. We found that BNT162b2 (vs CoronaVac) or booster doses (vs two doses) were significantly associated with higher serum IgG levels, but were not significantly associated with salivary IgA levels, regardless of prior infection status. Among non-infected individuals, serum IgG, serum IgA and salivary IgG levels were significantly higher against the ancestral strain than the Omicron BA.2 sublineage, but salivary IgA levels did not differ between the strains. Salivary IgA had the weakest correlation with serum IgG (r = 0.34) compared with salivary IgG (r = 0.63) and serum IgA (r = 0.60). Our findings suggest that intramuscular COVID-19 vaccines elicit a distinct mucosal IgA response that differs from the systemic IgG response. As mucosal IgA independently correlates with protection, vaccine trials should include mucosal IgA as an outcome measure.


Isolation and characterization of human H7N9 mAbs in vitro
a FACS depicting the staining and selection of H7-specific B cells from donor H7N9_HK2013 PBMCs. SSC-A, side scatter area; FSC-A, forward scatter area. b ELISA binding curves of the indicated mAbs to soluble recombinant H7N9 HA, with and without Endo H treatment, to H7N9 HA1 from 2013, 2016, and 2017, to H7N7 HA, and to 6 non-H7 HA or HA1. c Western blot of a cleaved H7 HA (molecular mass of 43 kDa for HA1 and 30 kDa for HA2) with mAb H7.HK2 or H7.HK4. d Neutralization curves of H7.HK mAbs against H7N9 2013 and 2017 pseudo viruses in MDCK cells. Data shown are mean ± SEM. Source data are provided in the Source Data file. Similar results were independently reproduced at least once.
Structural analysis of H7.HK1 and H7.HK2 in complex with H7 HA trimer
a Cryo-EM structures of H7.HK1 and H7.HK2 bound to H7 HA in the head region. b Top view of alignment of H7.HK1 and H7.HK2 complex structures. c Surface presentation of the H7.HK1 epitope (orange) on H7 HA1, with interacting CDRs shown. d H7.HK1 heavy chain forms seven hydrogen bonds and one salt bridge with H7 HA1. e H7.HK1 light chain forms one additional hydrogen bond with H7 HA1, and the interactions are stabilized by hydrophobic residues on the periphery of the light chain interface. f Modeling published structures of H7 HA1-binding antibodies (PDB: 6II4, 6II8, 6II9, 5V2A) onto the H7.HK1 bound structure, with an escape mutation R57K (green) reported for mAb 07-5F01. Competition ELISA with biotinylated H7.HK2 binding to the H7 HA monomer, in which unlabeled competing mAbs were titrated at increasing concentrations to evaluate the effect on H7.HK2 binding. Source data is provided in the Source Data file. g Sequence analysis of N = 1,483 H7 HA1s revealed a conserved lateral patch that largely overlaps with the H7.HK1 epitope. h Modeling the binding site of human receptor analogue LSTc (red) based on a previous crystal structure (PDB: 4BSE) onto H7 from the H7.HK1 complex, showing that H7.HK1 does not compete with sialic acid on the adjacent protomer (black). Alignment of the H7.HK1 complex with a previous crystal structure of H7 (PDB: 4BSE) shows that the 220-loop (pink) required for sialic acid binding (G218-G228) is disordered and would clash with the H7.HK1 light chain if it were present. Green asterisk symbol denotes the <2 Å clash between the CDR L1 N28 and the predicted location of P221 on HA1.
Prophylactic and therapeutic effects of human H7N9 mAbs in mice i.n. challenged with 10 LD50 of A/Anhui/1/2013 H7N9
a Female mice were i.p. injected 100 μg (equivalent of 5 mg/kg) or 20 μg (equivalent of 1 mg/kg) of the indicated mAbs (as human IgG1 unless otherwise specified) one day before viral challenge; % survival (<20% weight loss) and % body weight of survived mice were plotted over time. b Female mice were i.p. injected 100 μg of the indicated mAbs one day after viral challenge; % survival and % body weight of survived mice were plotted over time. Arrows indicate the time when mAbs were administered. Control groups of a non-H7 placebo mAb and PBS were included. Data for each group were combined from 1 to 2 experiments and shown as mean ‒ SEM. Source data with P values from two-sided unpaired student’s t-test are provided in the Source Data file. Asterisk symbols denote P < 0.05, and # denote P < 0.1.
H7.HK1 and two other lateral patch-binding antibodies define a conserved supersite of vulnerability on the HA head
a Representation of H7.HK1, Fab6649, and 045-09-2B05 bound to their respective HAs indicates diverse angles of approach and heavy/light chain orientations towards the lateral patch supersite. b Comparison of epitopes of H7.HK1, Fab6649, and 045-09-2B05 centering on the lateral patch defines the lateral patch supersite (blue). c A subset of epitope surface, centered on the lateral patch, overlaps between H7.HK1, Fab6649, and 045-09-2B05 (magenta). Of shared epitope surface, a subset of epitope residues is conserved (positions 121, 126, 168, and 172 by H3 numbering). d The four lateral patch antibodies contact conserved residues using diverse chemistry. Table displays the type of interaction between each residue and antibody.
Human neutralizing antibodies target a conserved lateral patch on H7N9 hemagglutinin head

May 2024

·

21 Reads

·

2 Citations

Avian influenza A virus H7N9 causes severe human infections with >30% fatality. Currently, there is no H7N9-specific prevention or treatment for humans. Here, from a 2013 H7N9 convalescent case in Hong Kong, we isolate four hemagglutinin (HA)-reactive monoclonal antibodies (mAbs), with three directed to the globular head domain (HA1) and one to the stalk domain (HA2). Two clonally related HA1-directed mAbs, H7.HK1 and H7.HK2, potently neutralize H7N9 and protect female mice from lethal H7N9/AH1 challenge. Cryo-EM structures reveal that H7.HK1 and H7.HK2 bind to a β14-centered surface and disrupt the 220-loop that makes hydrophobic contacts with sialic acid on an adjacent protomer, thereby blocking viral entry. Sequence analysis indicates the lateral patch targeted by H7.HK1 and H7.HK2 to be conserved among influenza subtypes. Both H7.HK1 and H7.HK2 retain HA1 binding and neutralization capacity to later H7N9 isolates from 2016–2017, consistent with structural data showing that the antigenic mutations during this timeframe occur at their epitope peripheries. The HA2-directed mAb H7.HK4 lacks neutralizing activity but when used in combination with H7.HK2 moderately augments female mouse protection. Overall, our data reveal antibodies to a conserved lateral HA1 supersite that confer neutralization, and when combined with a HA2-directed non-neutralizing mAb, augment protection.


Figure 5 In vivo activity of compound 172. (A) Permeability of compound 172 in Caco-2 cells and metabolic stability of 172 in mouse and human Liver microsomes. (B) In vivo pharmacokinetic profiling of compound 172 in a BALB/c mouse model (n Z 3). (C) Schematics of compound 172 administration in a golden Syrian hamster model. Two groups of hamsters (n Z 4) were given intranasal inoculations of 1000 PFU/animal of WT SARS-CoV-2. The treatment group (n Z 4) received compound 172 (1 mg/kg) or nirmatrelvir (200 mg/kg) via intraperitoneal (ip) injection, or pelitinib (10 mg/kg) via oral gavage. The vehicle group (n Z 4) was given 5% DMSO in 20% SBE-b-CD/0.9% saline. On the third day following the infection, the hamsters were euthanized for both viral yield and histopathological examination. (D) Hamsters lung and nasal turbinate viral yield were determined by RT-qPCR. Statistical analysis by one-way ANOVA: Data present as mean AE SD. ***P < 0.001; **P < 0.01; *P < 0.05. (E) Representative images of H&E-stained and IF-stained lung tissue sections from hamster treated as indicated. Scale bars: 100 mm. (F) Schematic of compound 172 administration in a K18-hACE2 mouse model. K18-hACE2 mice were intranasally inoculated with either Alpha (B.1.1.7, n Z 5 for survival rate) or Omicron (BA.5, n Z 4 for viral load detection) at 200 PFU/animal or 10,000 PFU/animal, respectively. The treatment group was given compound 172 (50 mg/kg) dissolved in a solution of 20% SBE-b-CD/0.9% saline, administered once daily via i.p. injection. The vehicle group was given 5% DMSO in 20% SBE-b-CD/0.9% saline using the same treatment regimen. The positive control group was given Nirmatrelvir (200 mg/kg) dissolved in a solution of 20% SBE-b-CD and 0.9% saline using the same treatment regimen. The mice in the survival study were monitored daily, and drugs were administered until they reached the humane endpoint or died. The mice in the viral load study were euthanized on the third day after the infection, and their lungs and nasal turbinate were collected for viral load quantification. (G) Mouse survival rate (upper panel) and daily body weight changes (lower panel) of K18-hACE2 mice. The comparison of survival rates between groups were analysed using Log-rank (ManteleCox) tests and that of body weight using two-way ANOVA. (H) K18-hACE2 mouse lung and nasal turbinate viral titer determined by standard plaque assay. Statistical analysis by one-way ANOVA: Data are presented as mean AE SD. ***P < 0.001; **P < 0.01; *P < 0.05; ns indicates not significant.
Identification of novel small-molecule inhibitors of SARS-CoV-2 by chemical genetics

May 2024

·

100 Reads

·

3 Citations

Acta Pharmaceutica Sinica B

There are only eight approved small molecule antiviral drugs for treating COVID-19. Among them, four are nucleotide analogues (remdesivir, JT001, molnupiravir, and azvudine), while the other four are protease inhibitors (nirmatrelvir, ensitrelvir, leritrelvir, and simnotrelvir-ritonavir). Antiviral resistance, unfavourable drug‒drug interaction, and toxicity have been reported in previous studies. Thus there is a dearth of new treatment options for SARS-CoV-2. In this work, a three-tier cell-based screening was employed to identify novel compounds with anti-SARS-CoV-2 activity. One compound, designated 172, demonstrated broad-spectrum antiviral activity against multiple human pathogenic coronaviruses and different SARS-CoV-2 variants of concern. Mechanistic studies validated by reverse genetics showed that compound 172 inhibits the 3-chymotrypsin-like protease (3CLpro) by binding to an allosteric site and reduces 3CLpro dimerization. A drug synergistic checkerboard assay demonstrated that compound 172 can achieve drug synergy with nirmatrelvir in vitro. In vivo studies confirmed the antiviral activity of compound 172 in both Golden Syrian Hamsters and K18 humanized ACE2 mice. Overall, this study identified an alternative druggable site on the SARS-CoV-2 3CLpro, proposed a potential combination therapy with nirmatrelvir to reduce the risk of antiviral resistance and shed light on the development of allosteric protease inhibitors for treating a range of coronavirus diseases.


Citations (76)


... Image processing and analysis can be complex, and resolution may be lower compared to X-ray crystallography for some samples. [134,135] Mass Spectrometry ...

Reference:

Mechanistic Insights into the Mutational Landscape of the Main Protease/3CLPro and Its Impact on Long-Term COVID-19/SARS-CoV-2 Management
Identification of novel small-molecule inhibitors of SARS-CoV-2 by chemical genetics

Acta Pharmaceutica Sinica B

... Jia and colleagues found that sialic acid binding is passively inhibited by their antibody, H7.HK1. This antibody inhibits the HA 220-loop (G218-G228 in H7 numbering, or G228-238 in H3 numbering), which makes hydrophobic contacts with sialic acid [62]. It is worth noting, however, that these mechanisms may not translate between influenza subtypes and further research is needed to understand how CL6649 and H7.HK1 compare. ...

Human neutralizing antibodies target a conserved lateral patch on H7N9 hemagglutinin head

... Our data show that the NA substitution from isoleucine to valine at position 223 and the change of serine by asparagine at position 247 have only a marginal effect on the susceptibility of A(H1N1)pdm09 viruses to NA inhibitors. However, NAs bearing both substitutions demonstrated a more than 10-fold reduction in susceptibility to oseltamivir and almost a four-fold reduction in susceptibility to zanamivir compared to wild-type virus [35]. Enzymatic analysis has proven that the NA-I223V substitution can offset the loss of affinity of NA for its substrate resulting from the NA-H275Y resistance substitution, while simultaneously boosting resistance [36]. ...

Global emergence of neuraminidase inhibitor-resistant influenza A(H1N1)pdm09 viruses with I223V and S247N mutations: implications for antiviral resistance monitoring
  • Citing Article
  • March 2024

The Lancet Microbe

... Cyclosporine A (CyA) is a classic immunosuppressant widely used to prevent organ transplant rejection and treat various autoimmune diseases, including rheumatoid arthritis and systemic lupus erythematosus. Its primary mechanism of action involves inhibiting calcineurin activity, which suppresses the synthesis and release of interleukin-2 (IL-2), thereby preventing T-cell activation and proliferation and achieving an immunosuppressive effect [16].In Finland, Koskela et al. conducted a study comparing the long-term prognosis of 62 pediatric Henoch-Schönlein purpura nephritis (HSPN) patients treated with methylprednisolone (MP) or CyA between 1996 and 2011, with an average follow-up of 10.8 years. Results showed that the CyA group had more sustained effects in reducing proteinuria and improving renal function, and the need for additional immunosuppressive therapy was significantly lower than in the MP group. ...

Nsp1 facilitates SARS-CoV-2 replication through calcineurin-NFAT signaling
  • Citing Article
  • February 2024

... Five hamsters were sampled in each group (as specified in the figure legends), and three sections from each animal were used for histology analysis. Histopathological scores were quantified using a semiquantitative method we previously described 35,52 . Table 4. ...

Divergent trajectory of replication and intrinsic pathogenicity of SARS-CoV-2 Omicron post-BA.2/5 subvariants in the upper and lower respiratory tract

EBioMedicine

... Additionally, soluble ACE2 (sACE2) proteins have been explored as antiviral inhibitors ( 22 -27 ), offering the advantages of broadly blocking antibody-escape variants of SARS-CoV-2 such as Delta and Omicron. However, it has been observed that low doses of sACE2 can facilitate SARS-CoV-2 infection ( 28 ), although this finding is still under debate ( 29 , 30 ). An outstanding question is whether soluble ACE2 or ACE2 mimetics can serve as viral receptors when associated with the cell membrane. ...

Soluble ACE2-mediated cell entry of SARS-CoV-2 via interaction with proteins related to the renin-angiotensin system
  • Citing Article
  • November 2023

Cell

... The inactivated vaccine regimens tested in this study induced both neutralizing responses and cytokine responses indicative of cellular immune responses in small animals. Given that immunization with monovalent I-P60 resulted in neutralizing responses against the original-D614G, delta, BA.1, and to some extent, BA.5 viruses, the I-P60 preserved an important feature reported for other inactivated vaccines based on original SARS-CoV-2, which also induce neutralizing responses against early omicron variants BA.1, BA.2, and BA.5 30,31 . Comparing neutralizing titers to those induced by commercially available vaccines across studies is challenged by differences in immunization regimens and neutralization assays. ...

Monovalent vaccination with inactivated SARS-CoV-2 BA.5 protects hamsters against Omicron but not non-Omicron variants
  • Citing Article
  • November 2023

npj Vaccines

... Whole genome sequencing was performed using Oxford Nanopore MinION device (Oxford Nanopore Technologies, Oxford, United Kingdom) as we described previously. [20][21][22] Nanopore sequencing was performed following the Nanopore protocol-PCR tiling of COVID-19 (Version: PTC_9096_v109_revH_06Feb2020) according to the manufacturer's instructions with minor modifications (Oxford Nanopore Technologies, Oxford, United Kingdom). Briefly, extracted RNA was first reverse transcribed to cDNA using SuperScript™ IV reverse transcriptase (Thermo Fisher Scientific, Waltham, Massachusetts, USA). ...

Humoral and cellular immunity against different SARS-CoV-2 variants in patients with chronic kidney disease

... TMPRSS2 expression 17,20 . Yet in the airway epithelium where abundant TMPRSS2 is expressed 16,21 , TMPRSS2-dependent virus entry remains as the dominant pathway utilized by human-pathogenic coronaviruses 3,[12][13][14][15][16]22,23 . In addition, TMPRSS2 is a key determinant that impacts coronavirus transmission and virus-induced tissue pathologies in the infected host [24][25][26] , suggesting the indispensable role of TMPRSS2 to coronavirus entry and pathogenesis at the primary infection sites. ...

The viral fitness and intrinsic pathogenicity of dominant SARS-CoV-2 Omicron sublineages BA.1, BA.2, and BA.5

EBioMedicine

... Monoclonal antibodies like the casirivimab/imdevimab combination bind to distinct epitopes within the spike protein receptor domain. This binding interferes with the virus's ability to interact with ACE2, thereby inhibiting the virus's intracellular penetration [37]. The Omicron variant, specifically its subvariant BA.2.75.2, has shown resistance to monoclonal antibodies and antiviral drugs [38] (Fig. 2). ...

Broad-spectrum humanized monoclonal neutralizing antibody against SARS-CoV-2 variants, including the Omicron variant