PUBLICATIONS

2024

Linn, Aung Khine, Suwimon Manopwisedjaroen, Phongthon Kanjanasirirat, Suparerk Borwornpinyo, Suradej Hongeng, Phetcharat Phanthong, and Arunee Thitithanyanont. (2024) 2024. “Unveiling the Antiviral Properties of Panduratin A through SARS-CoV-2 Infection Modeling in Cardiomyocytes”. International Journal of Molecular Sciences 25 (3). https://doi.org/10.3390/ijms25031427.

Establishing a drug-screening platform is critical for the discovery of potential antiviral agents against SARS-CoV-2. In this study, we developed a platform based on human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) to investigate SARS-CoV-2 infectivity, with the aim of evaluating potential antiviral agents for anti-SARS-CoV-2 activity and cardiotoxicity. Cultured myocytes of iPSC-CMs and immortalized human cardiomyocyte cell line (AC-16) were primarily characterized for the expression of cardiac markers and host receptors of SARS-CoV-2. An infectivity model for the wild-type SARS-CoV-2 strain was then established. Infection modeling involved inoculating cells with SARS-CoV-2 at varying multiplicities of infection (MOIs) and then quantifying infection using immunofluorescence and plaque assays. Only iPSC-CMs, not AC16 cells, expressed angiotensin-converting enzyme 2 (ACE-2), and quantitative assays confirmed the dose-dependent infection of iPSC-CMs by SARS-CoV-2, unlike the uninfectable AC16 cells lacking the expression of ACE2. Cytotoxicity was evaluated using MTT assays across a concentration range. An assessment of the plant-derived compound panduratin A (panA) showed cytotoxicity at higher doses (50% cytotoxic concentration (CC50) 10.09 μM) but promising antiviral activity against SARS-CoV-2 (50% inhibition concentration (IC50) 0.8-1.6 μM), suppressing infection at concentrations 10 times lower than its CC50. Plaque assays also showed decreased viral production following panA treatment. Overall, by modeling cardiac-specific infectivity, this iPSC-cardiomyocyte platform enables the reliable quantitative screening of compound cytotoxicity alongside antiviral efficacy. By combining disease pathogenesis and pharmacology, this system can facilitate the evaluation of potential novel therapeutics, such as panA, for drug discovery applications.

Purwono, Priyo Budi, Vimvara Vacharathit, Suwimon Manopwisedjaroen, Natali Ludowyke, Ampa Suksatu, and Arunee Thitithanyanont. 2024. “Infection kinetics, syncytia formation, and inflammatory biomarkers as predictive indicators for the pathogenicity of SARS-CoV-2 Variants of Concern in Calu-3 cells”. PLOS ONE 19 (4): 1-24. https://doi.org/10.1371/journal.pone.0301330.
The ongoing COVID-19 pandemic has led to the emergence of new SARS-CoV-2 variants as a result of continued host-virus interaction and viral genome mutations. These variants have been associated with varying levels of transmissibility and disease severity. We investigated the phenotypic profiles of six SARS-CoV-2 variants (WT, D614G, Alpha, Beta, Delta, and Omicron) in Calu-3 cells, a human lung epithelial cell line. In our model demonstrated that all variants, except for Omicron, had higher efficiency in virus entry compared to the wild-type. The Delta variant had the greatest phenotypic advantage in terms of early infection kinetics and marked syncytia formation, which could facilitate cell-to-cell spreading, while the Omicron variant displayed slower replication and fewer syncytia formation. We also identified the Delta variant as the strongest inducer of inflammatory biomarkers, including pro-inflammatory cytokines/chemokines (IP-10/CXCL10, TNF-α, and IL-6), anti-inflammatory cytokine (IL-1RA), and growth factors (FGF-2 and VEGF-A), while these inflammatory mediators were not significantly elevated with Omicron infection. These findings are consistent with the observations that there was a generally more pronounced inflammatory response and angiogenesis activity within the lungs of COVID-19 patients as well as more severe symptoms and higher mortality rate during the Delta wave, as compared to less severe symptoms and lower mortality observed during the current Omicron wave in Thailand. Our findings suggest that early infectivity kinetics, enhanced syncytia formation, and specific inflammatory mediator production may serve as predictive indicators for the virulence potential of future SARS-CoV-2 variants.
Huang, Yu-Ching, Sheng-Fan Wang, Bo-Cheng Chen, Zih-Syuan Yang, Meng-Chi Li, Xun-Ying Wu, Meng-Jey Youh, et al. 2024. “Towards cost-effective and lightweight surface plasmon resonance biosensing for H5N1 avian influenza virus detection: Integration of novel near-infrared organic photodetectors”. Sensors and Actuators B: Chemical 400: 134898. https://doi.org/https://doi.org/10.1016/j.snb.2023.134898.
H5N1 avian influenza virus (AIV) persists in causing highly fatal human infections, demanding rapid and accurate diagnostic assessment. In this study, we introduce an intensity-based SPR sensor utilizing NIR wavelength excitation in tandem with a specifically engineered NIR-OPD. The active layer of the OPD consists of a PTB7-Th and COTIC-4 F blend, offering an optimized response for a 980 nm excitation wavelength. Key performance metrics of this OPD include a low Jd of 0.185 nA/cm2, a high responsivity of 0.35 A/W, an EQE of 44.74%, and an exceptional detectivity of 4.59 × 1013 Jones at 980 nm wavelength under zero bias. It also exhibits a wide LDR of 113 dB. The integration of such OPDs into our SPR sensor provides advantages in compactness and cost-effectiveness. Employing this sensor, we detected the H5N1 AIV using a custom high-affinity polyclonal antibody against HA envelope of the H5N1 virus, completing analyses of culture medium samples within 12 min. The detection limit of this biosensor for the H5N1 AIV in PBS-diluted culture medium is approximately 4.3 × 104 copies/mL. When compared to a commercial H5-Ag lateral flow test kit, our biosensor showed a sensitivity 37 times higher. Key attributes of our biosensor include 3D printing technology for easy alignment of optical components and a rapid, simplified detection procedure. Collectively, our findings open up the potential of our SPR biosensor as an efficient tool for detecting H5N1 AIV, promising advancements in on-site detection methodologies.
Pisuttinusart, Nuttapat, Balamurugan Shanmugaraj, Chanya Srisaowakarn, Chutitorn Ketloy, Eakachai Prompetchara, Arunee Thitithanyanont, and Waranyoo Phoolcharoen. (2024) 2024. “Immunogenicity of a Recombinant Plant-Produced Respiratory Syncytial Virus F Subunit Vaccine in Mice”. Biotechnology Reports (Amsterdam, Netherlands) 41: e00826. https://doi.org/10.1016/j.btre.2023.e00826.

Respiratory syncytial virus (RSV) is a highly infectious respiratory virus that causes serious illness, particularly in young children, elderly people, and those with immunocompromised individuals. RSV infection is the leading cause of infant hospitalization and can lead to serious complications such as pneumonia and bronchiolitis. Currently, there is an RSV vaccine approved exclusively for the elderly population, but no approved vaccine specifically designed for infants or any other age groups. Therefore, it is crucial to continue the development of an RSV vaccine specifically tailored for these populations. In this study, the immunogenicity of the two plant-produced RSV-F Fc fusion proteins (Native construct and structural stabilized construct) were examined to assess them as potential vaccine candidates for RSV. The RSV-F Fc fusion proteins were transiently expressed in Nicotiana benthamiana and purified using protein A affinity column chromatography. The recombinant RSV-F Fc fusion protein was recognized by the monoclonal antibody Motavizumab specific against RSV-F protein. Moreover, the immunogenicity of the two purified RSV-F Fc proteins were evaluated in mice by formulating with different adjuvants. According to our results, the plant-produced RSV-F Fc fusion protein is immunogenic in mice. These preliminary findings, demonstrate the immunogenicity of plant-based RSV-F Fc fusion protein, however, further preclinical studies such as antigen dose and adjuvant optimization, safety, toxicity, and challenge studies in animal models are necessary in order to prove the vaccine efficacy.

Puthanakit, Thanyawee, Eakachai Prompetchara, Sivaporn Gatechompol, Chutitorn Ketloy, Arunee Thitithanyanont, Anan Jongkaewwattana, Supranee Buranapraditkun, et al. (2024) 2024. “Phase II Prefusion Non-Stabilised Covid-19 MRNA Vaccine Randomised Study”. Scientific Reports 14 (1): 2373. https://doi.org/10.1038/s41598-023-49653-6.

ChulaCov19 mRNA vaccine demonstrated promising phase 1 results. Healthy adults aged 18-59 years were double-blind randomised 4:1 to receive two intramuscular doses of ChulaCov19 50 µg or placebo. Primary endpoints were safety and microneutralization antibody against-wild-type (Micro-VNT50) at day 50. One hundred fifty adults with median (IQR) age 37 (30-46) years were randomised. ChulaCov19 was well tolerated, and most adverse events were mild to moderate and temporary. Geometric mean titres (GMT) of neutralizing titre against wild-type for ChulaCov19 on day 50 were 1367 IU/mL. T-cell IFN-γ-ELISpot showed the highest responses at one week (Day29) after dose 2 then gradually declined. ChulaCov19 50 µg is well tolerated and elicited high neutralizing antibodies and strong T-cell responses in healthy adults.Trial registration number: ClinicalTrials.gov Identifier NCT04566276, 28/09/2020.

2023

Niyomnaitham, Suvimol, Anan Jongkaewwattana, Atibordee Meesing, Nawamin Pinpathomrat, Sira Nanthapisal, Nattiya Hirankarn, Sarawut Siwamogsatham, et al. (2023) 2023. “Immunogenicity of a Fractional or Full Third Dose of AZD1222 Vaccine or BNT162b2 Messenger RNA Vaccine After Two Doses of CoronaVac Vaccines Against the Delta and Omicron Variants”. International Journal of Infectious Diseases : IJID : Official Publication of the International Society for Infectious Diseases 129: 19-31. https://doi.org/10.1016/j.ijid.2023.01.022.

OBJECTIVES: The study aimed to compare the immunogenicity and safety of fractional (half) third doses of heterologous COVID-19 vaccines (AZD1222 or BNT162b2) to full doses after the two-dose CoronaVac and when boosting after three different extended intervals.

METHODS: At 60-<90, 90-<120, or 120-180 days intervals after the two-dose CoronaVac, participants were randomized to full-dose or half-dose AZD1222 or BNT162b2, followed up at day 28, 60, and 90. Vaccination-induced immune responses to Ancestral, Delta, and Omicron BA.1 strains were evaluated by antispike, pseudovirus, and microneutralization and T cell assays. Descriptive statistics and noninferiority cut-offs were reported as geometric mean concentration or titer and concentration or titer ratios comparing baseline to day 28 and day 90 and different intervals.

RESULTS: No safety concerns were detected. All assays and intervals showed noninferior immunogenicity between full doses and half doses. However, full-dose vaccines and/or longer 120-180-day intervals substantially improved the immunogenicity (measured by antispike or measured by pseudotyped virus neutralizing titers 50; P <0.001). Seroconversion rates were over 90% against the SARS-CoV-2 strains by all assays. Immunogenicity waned more quickly with half doses than full doses but remained high against the Ancestral or Delta strains. Against Omicron, the day 28 immunogenicity increased with longer intervals than shorter intervals for full-dose vaccines.

CONCLUSION: Immune responses after day 28 when boosting at longer intervals after the two-dose CoronaVac was optimal. Half doses met the noninferiority criteria compared with the full dose by all the immune assays assessed.

Yang, Zih-Syuan, Chih-Yen Lin, Muhammad Bilal Khan, Ming-Cheng Hsu, Wanchai Assavalapsakul, Arunee Thitithanyanont, and Sheng-Fan Wang. (2023) 2023. “Understanding the Role of Galectins Toward Influenza A Virus Infection”. Expert Opinion on Therapeutic Targets 27 (10): 927-37. https://doi.org/10.1080/14728222.2023.2263912.

INTRODUCTION: Influenza A virus (IAV) is highly contagious and causes respiratory diseases in birds, mammals, and humans. Some strains of IAV, whether from human or avian sources, have developed resistance to existing antiviral drugs. Therefore, the discovery of new influenza antiviral drugs and therapeutic approaches is crucial. Recent studies have shown that galectins (Gal), a group of β-galactose-binding lectins, play a role in regulating various viral infections, including IAVs.

AREAS COVERED: This review provides an overview of the roles of different galectins in IAV infection. We discuss the characteristics of galectins, their impact on IAV infection and spread, and highlight their positive or negative regulatory functions and potential mechanisms during IAV infection. Furthermore, we explore the potential application of galectins in IAV therapy.

EXPERT OPINION: Galectins were first identified in the mid-1970s, and currently, 15 mammalian galectins have been identified. While all galectin members possess the carbohydrate recognition domain (CRD) that interacts with β-galactoside, their regulatory functions vary in different DNA or RNA virus infections. Certain galectin members have been found to regulate IAV infection through diverse mechanisms. Therefore, a comprehensive understanding of their roles in IAV infection is essential, as it may pave the way for novel therapeutic strategies.

Prompetchara, Eakachai, Chutitorn Ketloy, Mohamad-Gabriel Alameh, Kittipan Tharakhet, Papatsara Kaewpang, Nongnaphat Yostrerat, Patrawadee Pitakpolrat, et al. (2023) 2023. “Immunogenicity and Protective Efficacy of SARS-CoV-2 MRNA Vaccine Encoding Secreted Non-Stabilized Spike in Female Mice”. Nature Communications 14 (1): 2309. https://doi.org/10.1038/s41467-023-37795-0.

Establishment of an mRNA vaccine platform in low- and middle-income countries (LMICs) is important to enhance vaccine accessibility and ensure future pandemic preparedness. Here, we describe the preclinical studies of "ChulaCov19", a SARS-CoV-2 mRNA encoding prefusion-unstabilized ectodomain spike protein encapsulated in lipid nanoparticles (LNP). In female BALB/c mice, ChulaCov19 at 0.2, 1, 10, and 30 μg elicits robust neutralizing antibody (NAb) and T cell responses in a dose-dependent relationship. The geometric mean titers (GMTs) of NAb against wild-type (WT, Wuhan-Hu1) virus are 1,280, 11,762, 54,047, and 62,084, respectively. Higher doses induce better cross-NAb against Delta (B.1.617.2) and Omicron (BA.1 and BA.4/5) variants. This elicited immunogenicity is significantly higher than those induced by homologous CoronaVac or AZD1222 vaccination. In a heterologous prime-boost study, ChulaCov19 booster dose generates a 7-fold increase of NAb against Wuhan-Hu1 WT virus and also significantly increases NAb response against Omicron (BA.1 and BA.4/5) when compared to homologous CoronaVac or AZD1222 vaccination. Challenge studies show that ChulaCov19 protects human-ACE-2-expressing female mice from COVID-19 symptoms, prevents viremia and significantly reduces tissue viral load. Moreover, anamnestic NAb response is undetectable in challenge animals. ChulaCov19 is therefore a promising mRNA vaccine candidate either as a primary or boost vaccination and has entered clinical development.

Chanprapaph, Kumutnart, Chutima Seree-aphinan, Ploysyne Rattanakaemakorn, Cherrin Pomsoong, Yanisa Ratanapokasatit, Chavachol Setthaudom, Arunee Thitithanyanont, et al. (2023) 2023. “A Real-World Prospective Cohort Study of Immunogenicity and Reactogenicity of ChAdOx1-S[recombinant] Among Patients With Immune-Mediated Dermatological Diseases”. The British Journal of Dermatology 188 (2): 268-77. https://doi.org/10.1093/bjd/ljac045.

BACKGROUND: Immunogenicity and reactogenicity of COVID-19 vaccines have been established in various groups of immunosuppressed patients; however, studies involving patients with immune-mediated dermatological diseases (IMDDs) are scarce.

OBJECTIVES: To investigate the influence of IMDDs on the development of SARS-CoV-2-specific immunity and side-effects following ChAdOx1-S[recombinant] vaccination.

METHODS: This prospective cohort study included 127 patients with IMDDs and 97 participants without immune-mediated diseases who received ChAdOx1-S[recombinant]. SARS-CoV-2-specific immunity and side-effect profiles were assessed at 1 month postvaccination and compared between groups. Immunological (primary) outcomes were the percentages of participants who tested positive for neutralizing antibodies [seroconversion rate (SR)] and those who developed T-cell-mediated immunity demonstrated by an interferon-γ-releasing assay (IGRA) [positive IGRA rate (+IGRA)]. Reactogenicity-related (secondary) outcomes were the unsolicited adverse reactions and worsening of IMDD activity reflected by the uptitration of immunosuppressants during and within 1 month of vaccination.

RESULTS: Overall, the SR for the IMDD group was similar to that of participants without immune-mediated conditions (75·6 vs. 84·5, P = 0·101), whereas + IGRA was lower (72·4 vs. 88·7, P = 0·003). Reactogenicity was similar between groups. No severe adverse reaction was reported. By stratifying the participants in the IMDD group according to individual disease, the immunogenicity of the vaccine was lowest in patients with autoimmune bullous diseases (AIBD) (SR 64·5%, +IGRA 62·9%) and highest in patients with psoriasis (SR 87·7%, +IGRA 80·7%). The reverse trend was found for vaccine-related reactions. Immunosuppressants were uptitrated in 15·8% of cases; 75% of these were patients with AIBD.

CONCLUSIONS: Among participants with IMDDs, ChAdOx1-S[recombinant] showed good immunogenicity among patients with psoriasis, but demonstrated lower levels of immunogenicity for patients with AIBD. Some patients, especially patients with AIBD, should be closely monitored as they may require treatment escalation within 1 month postvaccination.