top of page

CVVT x HKU
"2nd Symposium on Combating 
Emerging Infectious Diseases"

Date: 16 Dec 2022 
Time: 9:00am to 1:00pm
Venue: Lecture Theatre 2, G/F, William MW Mong Block, Faculty of Medicine Building, 21 Sasson Road

2nd symposium on CEID - Promotional Poster (Final).png

Registration is now closed. 
Thank you for your interest and overwhelming participation!

Program Rundown

Rundown of 2nd Symposium on Combating Emerging Infectious Diseases (Finalized)_20221214_Pa
Rundown of 2nd Symposium on Combating Emerging Infectious Diseases (Finalized)_20221214_Pa

Keynote Speakers

Prof David Ho

Prof Xiaoning Xu

Speakers

Prof HL Chen

Prof ZW Chen

Dr Kelvin To

Dr Jasper Chan

Dr Richard Kao

Dr Philip Yeung

Dr Raven Kok

Dr Hin Chu

Dr Jane Zhou

Dr SF Yuan

Poster Presenters

Dr Peng Gao

Dr Cun Li

Dr Li Liu

Dr Zhiwu Tan

 

Dr Bingpeng Yan & Dr Kong Hung Sze

Dr Pui Wang

Dr Anna Zhang

Dr Hanjun Zhao

 

Abstracts for Oral Presentation

Name & Affiliation:

Prof David Da-I Ho

Columbia University

 

Title:

How SARS-CoV-2 Evades Neutralizing Antibodies and Antiviral Drugs

 

Abstract:

This presentation will describe work done in the past two years on how SARS-CoV-2 has evolved in its spike protein to evade the action of antibodies elicited by Covid-19 infection or vaccination, and how this viral evolution has resulted in new variants that have come to dominate the pandemic in successive waves.  The extreme antibody evasion by most recent studies on novel variants such as BQ.1.1 and XBB will be reported.  Likewise, how SARS-CoV-2 escape from the action of nirmatrelvir, a main protease inhibitor, will be described, including the multiple pathways the virus could take to become resistant to this drug.

 

Biography:

Prof Ho is the founding Scientific Director and Chief Executive Officer of the Aaron Diamond AIDS Research Center, a world-renowned biomedical research institute. He is also the Clyde & Helen Wu Professor of Medicine, as well as a Professor of Microbiology & Immunology at Columbia University.

 

Prof Ho received his degrees from California Institute of Technology (1974) and Harvard Medical School (1978). Subsequently, he did his clinical training in internal medicine and infectious diseases at UCLA School of Medicine (1978-1982) and Massachusetts General Hospital (1982-1985), respectively. Prof Ho has been actively engaged in AIDS research for 40 years and has published over 400 papers on the subject. Among an impressive list of seminal contributions to the field, he is perhaps most recognized for the elucidation of the dynamic nature of HIV replication in infected persons. This basic understanding led Prof Ho and his coworkers to champion combination antiretroviral therapy that has resulted in dramatic reductions in AIDS-associated mortality since 1996. Prof Ho continues to pursue therapeutic studies that attempt to eradicate HIV. In addition, his research team is now devoting considerable efforts to develop a vaccine to halt the spread of the AIDS epidemic.  Since the start of the Covid-19 pandemic, his team has devoted considerable efforts to understand the causative agent, SARS-CoV-2.

Anchor 1
Anchor 2

Name & Affiliation:

Prof Xiaoning Xu

Imperial College London

Title:

Characterization of CoV-bnMABs: Implications for universal immunotherapy and vaccine development

Abstract:

Broadly neutralizing antibodies to coronaviruses (CoV-bnMABs) are invaluable reagents for the treatment of diverse coronaviruses infection as well as guidance of a universal vaccine design. To achieve this goal, we have isolated and characterized a human monoclonal antibody (OX24) from a patient recently infected by COVID-19. OX24 can bind to a conserved epitope presented in S2 domain of the spike proteins, and neutralise almost all human CoV isolates in vitro assays including the CoV-19 variants. In addition, OX24 shows a protective efficacy (prophylactic or therapeutic) against SARS-CoV-2 challenge in an animal model as indicated less weight loss and reduced lung pathology. Overall, OX24 provides effective tools against emerging pandemic human CoV of the future as well as the development of universal vaccines.

 

Biography:

Prof Xu is the Chair Professor in Human Immunology at Imperial College London. Over the last two decades, Prof Xu served as Medical Research Council (MRC) Senior Clinical Scientist, project leader, and tenured programme leader at MRC Human Immunology Unit, Oxford University, where he led research projects involving immune correlates of protection against emerging infectious diseases including HIV, SARS, and avian influenza. In 2010, he joined Novartis as the Head of Novartis Vaccines Research China, Novartis Vaccines & Diagnostics based in Shanghai.

Name & Affiliation:

Prof Honglin Chen

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

An intranasal influenza virus-vectored vaccine system for blocking SARS-CoV-2 and influenza virus infection

 

Abstract:

We have developed an intranasal vaccine candidate based on a live attenuated influenza virus (LAIV) with a deleted NS1 gene that encodes cell surface expression of the receptor-binding-domain (RBD) of the SARS-CoV-2 spike protein, designated DelNS1-RBD LAIV. Immune responses and protection against SARS-CoV-2 variants challenge following intranasal administration of DelNS1-RBD vaccines were analyzed in animal models and compared with intramuscular injection of the mRNA vaccine. Notably, vaccination with DelNS1-RBD4N-DAF LAIVs, but not mRNA vaccine, blocked replication of SARS-CoV-2 variants, including Delta and Omicron variants, in the respiratory tissues of animals.  A phased III study found DelNS1-RBD LAIV is highly safe and effective in humans.  More importantly, DelNS1-RBD LAIV retain the full ability to induce immunity against influenza viruses. The DelNS1-RBD LAIV system is an ideal technology for creating dual function vaccines against both influenza and COVID-19 for use in annual vaccination strategies.

 

Biography:

Prof. Honglin Chen research areas focus on molecular virology of emerging influenza and coronaviruses, with specific aims to explore molecular determinants for cross species transmission of avian influenza viruses and coronavirus. His research on biology of the NS1 protein of influenza virus has led to the establishment of a unique DelNS1 live attenuated influenza vaccine platform which can be used to develop vaccines for prevention and treatment of infection of respiratory viral diseases, including influenza and COVID-19.

Anchor 3

Name & Affiliation:

Prof Zhiwei Chen

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Protection against SARS-CoV-2 infection by antibody and T cell responses

 

Abstract:

Acute SARS-CoV-2 infection rapidly resulted in impaired and imbalanced host immune responses, which were associated with COVID-19 pathogenesis. During the natural course of SARS-CoV-2 infection, our findings on impaired dendritic and T cells together with over-activated extrafollicular B cells indicated the importance of balanced immune responses for vaccine-induced protective immunity. Vaccine-induced neutralizing antibody responses, however, waned significantly among vaccinees especially against SARS-CoV-2 variants of concern. Besides neutralizing escape viral variants, we found that robust nasal SARS-CoV-2 infection could outcompete with passive and vaccine-induced neutralizing antibodies at the portal of viral entry, underlying the increased number of vaccine-breakthrough infections and reinfections. We then demonstrated that booster vaccination through the intranasal route was essential for eliciting improved protection against intranasal viral infection. In addition, we found that vaccine-breakthrough infection elicited broadly neutralizing antibodies primarily targeted at two distinct domains in SARS-CoV-2 spike protein. Collectively, our findings contributed significantly to understanding human immune responses against SARS-CoV-2 infection.

 

Biography:

Prof. Zhiwei Chen is the director of AIDS Institute and tenured full professor of Department of Microbiology at the University of Hong Kong Li Ka Shing Faculty of Medicine. He did his postgraduate studies at Aaron Diamond AIDS Research Center (ADARC) and obtained his Ph.D. degree in Medical Microbiology from New York University School of Medicine, USA, in 1996.  He progressed from a post-doc to a research scientist, and then to a staff investigator/assistant professor all at ADARC of the Rockefeller University from 1996 to 2003.  He has joined the University of Hong Kong as the founding director of the AIDS Institute since 2007. He has focused his research on vaccine and immunotherapy against AIDS, SARS, COVID-19 and cancer with over 150 SCI publications. He received numerous research grants as the principal investigator from NIHR32, amFAR, NIHR01, Gates Foundation in USA; Wellcome funds in UK; RGC TRS/CRF/GRF, HMRF and ITF in Hong Kong, as well as a project leader of 973, 11th- and 12th- Mega grants in mainland China. He is one of the 2022 highly cited researchers by Clarivate Analytics.

Anchor 4

Name & Affiliation:

Dr Raven Kin Hang Kok

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Intradermal Influenza vaccine: combining intradermal route of administration and use of a rationally designed single-round infectious influenza virus

 

Abstract:

Influenza causes millions of infections yearly despite the availability of vaccines. Here, we describe the development and characterization of a novel live attenuated IntraDermal Influenza Vaccine, IDIV, a regimen combining the use of a rationally designed single-round infectious virus and intradermal route of administration. Third generation SMRT long-read sequencing was first applied for the rational design of prototypic IDIV, which was then productively generated using a trans-complementation system. Single-dose intradermal administration of IDIV without adjuvant in mice elicited potent innate/adaptive immunity and high-titer neutralizing antibody production. In addition, IDIV elicits cross-protection against heterologous H1N1, H5N1 and H7N9. Taken together, this novel vaccination strategy, combining the intradermal route of administration and the use of a rationally designed single-round infectious influenza virus, elicits potent antibody response and heterologous influenza protection.

Biography:

Dr. Kok is currently an Associate Professor in the Department of Microbiology, The University of Hong Kong. He is a molecular virologist and has been interested in the host-virus interaction and host antiviral responses including “interferon signaling”. He first identified a host antiviral protein, PACT, that can recognize the viral defective interfering RNAs and optimally induce the production of type-I interferon, an indispensable primary antiviral response during early infection. He continued to characterize this novel antiviral mechanism and the viral interferon antagonists encoded by influenza A virus, Herpes Simplex virus, Measles virus and MERS-CoV and SARS-CoV-2. In addition to basic molecular virology study, Dr. Kok and his team recently rationally designed and developed novel vaccines such as intradermal influenza vaccine, nasal COVID-19 vaccine protein booster and single-rounded live SARS-CoV2 vaccine.

Anchor 5

Name & Affiliation:

Dr Richard Yi-Tsun Kao

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Viral nucleoprotein (NP) as a druggable target for influenza virus antiviral therapy

 

Abstract:

Influenza A viruses (IAVs) continue to pose an imminent threat to mankind. The suboptimal vaccine coverage and efficacy, coupled with recurrent events of viral resistance against a very limited antiviral portfolio, emphasize an urgent need for new additional prophylactic and therapeutic options. Since our identification of influenza viral nucleoprotein (NP) as an effective druggable target and nucleozin a potent antagonist of NP with an novel mode of action, a number of research groups have used NP as a target for antiviral drug development. We have recently characterized yet another novel NP inhibitor FA-6005 which is structurally different from nucleozin and with a totally different mode of action. There are two FA-6005 binding sites in the NP from the X-ray crystallography studies with one binding site partially overlapping with the known nucleozin binding sites. A number of naturally occurring and artificially induced mutations are involved in defining the binding mode of these two novel NP inhibitors.

Biography:

Dr. Kao received his PhD in Microbiology in 1999 from UBC under the supervision of Professor Julian Davies and subsequent postdoctoral training at Harvard Medical School from 1999-2001. He joined the University of Hong Kong in 2001 and is now a tenured Associate Professor in the Department of Microbiology, School of Clinical Medicine,  Li Ka Shing Faculty of Medicine. Dr. Kao’s research focuses on the application of chemical genetics in infectious diseases, especially emerging and re-emerging viral and bacterial infections. His work on SARS-CoV has established the world’s first model of viral chemical genetics and illustrated that chemical genetic approach could be employed to probe druggable targets of a pathogenic virus. Dr. Kao’s team also employed similar approach to identify influenza A nucleoprotein as a novel druggable antiviral target and nucleozin a potent antagonist of the nucleoprotein. Most recently, Dr. Kao has extended his chemical genetic studies to bacterial virulence and antibiotic resistance and has illustrated the potential use of anti-virulence compounds to treat MRSA infections. Dr. Kao received the Innovation Academy Award from International Consortium of Prevention and Control of Infection (ICPIC) in Geneva, Switzerland in 2017 and recently the 2019 State Scientific and Technological Progress Award (Second-class).

Anchor 6

Name & Affiliation:

Dr Kelvin Kai-Wang To

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Genomic and serological surveillance of COVID-19

 

Abstract:

The Omicron variant has become the dominant SARS-CoV-2 viral lineage globally in 2022. Due to the numerous mutations in the spike protein, the Omicron variant was able to evade from neutralizing antibodies that were elicited from prior vaccination or infection. As a result, the Omicron variant was associated with a poorer vaccine efficacy and higher reinfection rate. In Hong Kong, the Omicron variant caused the severe fifth wave in early 2022, and our serosurveillance has revealed the lack of population immunity against the Omicron variant before this outbreak. In this talk, I will discuss about our COVID-19 genomic and serological surveillance studies. In particular, I will explore how we use surveillance data to predict the severity of upcoming COVID-19 waves.

 

Biography:

Dr. Kelvin To is currently the Department Chairperson and a Clinical Associate Professor of the Department of Microbiology, LKS Faculty of Medicine, the University of Hong Kong. His research focuses on emerging respiratory tract infection. Trained as a clinical microbiologist, he is particularly interested in studies which have direct impact on patient care or public health policies. During the COVID-19 pandemic, he has pioneered the use of saliva for diagnostic testing, described the antibody kinetics during acute and convalescent period, and used genomic and serological surveillance to assess the risk of emerging SARS-CoV-2 variants.

Anchor 7

Name & Affiliation:

Dr Jasper Fuk-Woo Chan

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

COVID-19: What have we learned from animal models?

 

Abstract:

Animal models are essential tools for investigating the pathogenesis, transmission, and countermeasures for emerging infectious diseases. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a lineage B betacoronavirus first discovered in Wuhan, China in late 2019, and the causative agent of the coronavirus disease 2019 (COVID-19) pandemic. Since the early phase of the pandemic, various animal models have been developed to help answering diverse questions of this emerging global health threat. The golden Syrian hamster model is a readily available small animal model that closely mimics many aspects of human COVID-19. In this session, I will discuss about how we established and applied the golden Syrian hamster model to enhance our understanding on COVID-19.

Biography:

Dr. Jasper Fuk-Woo Chan is a tenured Clinical Associate Professor at the Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong. His research focuses on the diagnosis, treatment, and control of emerging viral infections with pandemic potential and emerging infectious disease with special relevance to Asia. He has authored more than 320 publications in these areas, including first or corresponding-authored articles in The Lancet and Science, and has been ranked by Clarivate as a Highly Cited Researcher since 2021. He has served as expert member or ambassador of various international organizations, including as member of the WHO ad hoc Expert Group focused on COVID-19 disease modelling (WHOCOM) and as Young Ambassador of Science of the American Society for Microbiology. He and his team reported the world’s first familial cluster of COVID-19 that confirmed person-to-person transmission of SARS-CoV-2, which resulted in major policy changes for controlling the pandemic worldwide. His team also established the world’s first Syrian hamster model for COVID-19, which has now become one of the most commonly used animal models for COVID-19 research.

Name & Affiliation:

Dr Philip Man Lung Yeung

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Soluble ACE2: Friend or foe

 

Abstract:

In the past, it is unclear that why only a small population of the lung cells has a detectable expression level of cACE2 despite they are the major target cells of SARS-CoV-2. While most studies focus on the function of cACE2 in SARS-CoV-2 pathogenesis, the role of sACE2 is largely unknown. In this study, we have discovered a novel cell entry mechanism mediated by sACE2, explaining why SARS-CoV-2 can attack many organs, including lung, where cACE2 are poorly expressed. Our findings could potentially explain the viral tropism of SARS-CoV-2.  Overall, we also have provided experimental proves to show the importance but overlooked role of sACE2 in SARS-CoV-2 infection. The findings could shed new light on our understanding of pathogenesis and treatment strategies for COVID-19.

 

Biography:

Dr. Yeung is an Assistant Professor at Department of Microbiology, School of Clinical Medicine, HKU. His research focuses on studying the molecular pathogenesis and dissecting host-virus interaction in human pathogenic viruses using different cutting-edge technology platforms such as genome-wide loss-of-function (CRISPR/RNAi) screening, high-throughput sequencing, and transgenic mouse model. Using these state-of-the-art platforms, Dr. Yeung has been consistently producing high quality research works leading to publication in many international prestigious journals, including Cell, Nature Microbiology, Journal of Clinical Investigations, Nucleic Acids Research, and EMBO Reports. His recent work on SARS-CoV-2 has uncovered a novel cell entry mechanism of SARS-CoV-2 via interaction with soluble ACE2 (Cell, 2021). The findings broke new grounds and will substantially advance the field. His outstanding research achievement s have made him awarded the State Scientific and Technological Progress Award, the China Medical Science and Technology Award and the Faculty Outstanding Research Output Award.

Name & Affiliation:

Dr Hin Chu

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Evolutionary trajectory of the virological features and intrinsic pathogenicity of SARS-CoV-2 Omicron sublineages

 

Abstract:

Omicron remains as the dominant SARS-CoV-2 variant. Among the numerous Omicron sublineages that have emerged, BA.1, BA.2, BA.5, and their related sublineages have resulted in the largest number of infections. While recent studies demonstrated that these Omicron sublineages robustly escape neutralizing antibody response, it remains unclear on whether these Omicron sublineages share any pattern of evolutionary trajectory on their intrinsic replication efficiency and pathogenicity along the respiratory tract. Here, we investigated the virological features, replication capacity in the nasal epithelium and lung, and pathogenicity of BA.1, BA.2, BA.5. Our results indicated that the sequentially emerged Omicron sublineages are gaining intrinsic replication fitness in the nasal epithelium. In contrast, the Omicron sublineages are increasingly attenuated in lungs of infected K18-hACE2 and C57B6 mice, leading to decreased pathogenicity. Nevertheless, lung manifestations remain severe in Omicron sublineages-infected aged C57B6 and A129 mice, highlighting the importance of vaccination to the highly susceptible populations.

 

Biography:

Dr. Chu focuses on investigating the host and viral determinants and pathogenesis of emerging viral infections, with an emphasis on highly pathogenic coronaviruses. He has authored >130 publications with >50 of them as first/co-first or corresponding author (Google Scholar: h-index 46; Citation 24,789). According to Clarivate Analytics' Essential Science Indicators, Dr. Chu is ranked as one of the world's top 1% scholars by citations and as a "Highly Cited Researcher" in 2021 and 2022. His research is well supported by competitive grants including RGC-GRF, HMRF, NSFC, RGC-CRF, TRS, and Health@InnoHK. As a recognition of Dr. Chu’s track record on coronaviruses, he was awarded the NSFC-Excellent Young Scientist Fund (Hong Kong and Macau) in 2021.

Name & Affiliation:

Dr Jane Jie Zhou

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Establishing human respiratory organoids for studying SARS-CoV-2 infection

 

Abstract:

Conventionally, in vitro studies of human biology and pathology, including virus-host interaction, have hinged on the immortalized and cancer-derived cell lines that comprise homogenous clonal cells. These cell lines can barely model native human epithelia which are composed of multiple cell populations. Thus, a biologically relevant and readily available in vitro model system is desperately needed.

We have established a two-phase, bipotential organoid culture system that enables the generation of both types of human respiratory epithelium in culture plates. We derive organoids from primary lung tissues and nasal cells highly efficiently, which provide a stable and self-renewable source for long-term expansion. We then induced proximal and distal differentiation in the long-term expandable organoids and generated mature airway and alveolar organoids that morphologically and functionally phenocopy the native airway and alveolar epithelium, respectively (PNAS 2018, Cell Discovery 2022). Overall, we have established a bipotential organoid culture system able to reproducibly expand the entire human respiratory epithelium in vitro for modeling respiratory diseases, including influenza and COVID-19.

Biography:

Dr. Zhou obtained her PhD degree in University of Hong Kong in 2007. After her postdoctoral training in UC San Francisco, she moved back to HKU. Now she is an assistant professor in Department of Microbiology. Prior to her research career, she graduated from Xi An Medical University and served as a clinical pathologist in affiliated hospitals of Peking University.  Her research interest is focused on virus-host interaction and pathogenesis of viral infection. Her interdisciplinary background enables her to seamlessly integrate organoid technology into virus research.

Name & Affiliation:

Dr Shuofeng Yuan

Centre for Virology, Vaccinology and Therapeutics / The University of Hong Kong

 

Title:

Antiviral discovery against SARS-CoV-2

Abstract:

As the COVID-19 pandemic threatens is a long-term problem and vaccines do not promise complete and lasting protection, antiviral molecules will remain an important line of defense. Therefore, “vaccine + drug” strategy may hold the only promise to end the Pandemic. We profiled a library of drugs encompassing approximately 12,000 clinical-stage or Food and Drug Administration (FDA)-approved small molecules to identify candidate therapeutic drugs for COVID-19. We report the identification of clofazimine, an anti-leprosy drug with a favorable safety profile, possesses inhibitory activity against several coronaviruses. Metal compounds are historic antimicrobial agents; however, their antiviral activities have rarely been explored. In another study, we discovered that ranitidine bismuth citrate (Pylorid), a commonly used drug with a safe and comprehensive pharmacological profile, potently inhibited SARS-CoV-2 replication in vitro and in vivo. Pylorid is a potent irreversible inhibitor of SARS-CoV-2 helicase, which binds to SARS-CoV-2 helicase and releases zinc ions from zinc-binding domain. Taken together, our data provide evidence that clofazimine and Pylorid may have a role in the control of the current pandemic of COVID-19 and—possibly more importantly—in dealing with coronavirus diseases that may emerge in the future.

Biography:

Dr. Shuofeng Yuan is currently an Assistant Professor in HKU and a Visiting Investigator in Scripps Research Institute (USA). He is also an awardee of HMRF Research Fellowship by HKSAR Government. Dr. Yuan’s research focus on molecular virology and particularly host-targeted antiviral research. He makes the conceptual breakthrough which defines the role of host sterol regulatory element-binding protein (SREBP) and host AP-2 complex subunit mu (AP2M1) with broad relevance of different viruses’ replication. Dr. Yuan is also the key author of several high-quality research papers that represent major breakthroughs in SARS-CoV-2 and COVID-19 research. These papers published in Nature, Science and Lancet. His pioneering work has derived new targets, new strategies and novel lead compounds for antiviral therapy against SARS-CoV-2. He is the worldwide “Highly Cited Researchers” by Clarivate (for consecutive years 2021 and 2022).

Anchor 8
Anchor 9
Anchor 10
Anchor 11
Anchor 12

Abstracts for Poster Presentation

Anchor 13

Name: Dr Peng Gao

Title: Antivirulence Agent as an Adjuvant of Antibiotics in Treating Staphylococcal Infections

Abstract: Antibiotics are widely applied to treat infectious diseases. Empirically treatment with incorrect antibiotics, or even correct antibiotics always falls into subinhibitory concentrations, due to dosing, distribution, or secretion. In this study, we have systematically evaluated in vitro virulence induction effect of antibiotics and in vivo exacerbated infection. The major highlight of this work is to prove the β-lactam and tetracyclines antibiotics exacerbated disease is due to their induction effect on staphylococcal virulence. The combination of antivirulence agents and antibiotics can be a novel approach to controlling antibiotic-induced S. aureus pathogenicity. This phenomenon is common and suggests that if β-lactam antibiotics remain the first line of defense during empirical therapy, we either need to increase patient reliability or the treatment approach may improve in the future when paired with anti-virulence drugs. Our findings from this study demonstrated the benefits of antivirulence-antibiotic combinatorial treatment against S. aureus infections and provide a new perspective on the development of antibiotic adjuvants.

Name: Dr Cun Li

Title: Human Nasal Organoids Model SARS-CoV-2 Upper Respiratory Infection and Recapitulate the Differential Infectivity of Emerging Variants

Abstract: The human upper respiratory tract, specifically the nasopharyngeal epithelium, is the entry portal and primary infection site of respiratory viruses. Productive infection of SARS-CoV-2 in the nasal epithelium constitutes the cellular basis of viral pathogenesis and transmissibility. Yet a robust and well-characterized in vitro model of the nasal epithelium remained elusive. Here we report an organoid culture system of the nasal epithelium. We derived nasal organoids from easily accessible nasal epithelial cells with a perfect establishment rate. The derived nasal organoids were consecutively passaged for over 6 months. We then established differentiation protocols to generate 3-dimensional differentiated nasal organoids and organoid monolayers of 2-dimensional format that faithfully simulate the nasal epithelium. Notably, the differentiated nasal organoid monolayers accurately recapitulated higher infectivity and replicative fitness of the Omicron variant than the prior variants. SARS-CoV-2, especially the more transmissible Delta and Omicron variants, destroyed ciliated cells and disassembled tight junctions, thereby facilitating virus spread and transmission. In conclusion, we establish a robust organoid culture system of the human nasal epithelium for modeling upper respiratory infections and provide a physiologically-relevant model for assessing the infectivity of SARS-CoV-2 emerging variants.

Name: Dr Li Liu

Title: Δ42PD-1 suppresses BCR-mediated B cell activation in HIV-1 infection

Abstract: HIV-1 antibody responses are drained into exhausted B cells, yet the suppressor of B cell activation after B cell receptor (BCR) stimulation remains incompletely understood. Here, we report that Δ42PD-1, an isoformic PD-1, is predominantly up-regulated on activated memory, anergic naïve and tissue like memory B cells compared with PD-1 and FcRL4 in chronic HIV-1 patients (CHPs). BCR-stimulation up-regulates Δ42PD-1, resulting in B cell exhaustion, cell cycle arrest and death. Mechanistically, Δ42PD-1 recruits Src homology region 2 domain containing phosphatase 1 (SHP1) via its intracellular immunoreceptor tyrosine-based switch motif (ITSM). SHP1 then binds and inhibits AKT1 activation and thereby suppresses the AKT1/FOXO1 pathway. Δ42PD-1-specific antibody, however, reduces the SHP1 recruitment, increasing the AKT1/FOXO1 activation and proliferation of B cells derived from CHPs. Our findings demonstrate that Δ42PD-1 is a previously unrecognized suppressor of BCR-mediated B cell activation, which may serve as a potential immunotherapeutic target for restoring B cell functionality.

 

Name: Dr Zhiwu Tan

Title: Isoformic PD-1-mediated Immunosuppression Underlies Resistance to PD-1 Blockade in Hepatocellular Carcinoma Patients

Abstract: Immune checkpoint blockade (ICB) has improved cancer treatment, yet why most hepatocellular carcinoma (HCC) patients are resistant to PD-1 ICB remains elusive. Here, we investigated 3 cohorts of 74 HCC patients, including 41 untreated, 28 treated with Nivolumab and 5 treated with Pembrolizumab. We found distinct T cell subsets, which did not express PD-1 but expressed the isoform Δ42PD-1, accounting for up to 71% of cytotoxic T lymphocytes. Δ42PD-1+ T cells were tumor-infiltrating and correlated positively with HCC severity. Moreover, they were more exhausted than PD-1+ T cells by single T cell and functional analysis. HCC patients treated with PD-1 ICB showed effective PD-1 blockade but increased frequencies of Δ42PD-1+ T cells over time. Tumor-infiltrated Δ42PD-1+ T cells likely sustained HCC through TLR4-signaling for tumorigenesis. Anti-Δ42PD-1 antibody, but not Nivolumab, inhibited tumor growth in three HCC humanized mouse models. Our findings not only revealed a mechanism underlying resistance to PD-1 ICB but also identified anti-Δ42PD-1 antibody for HCC immunotherapy.

Name: Dr Bingpeng Yan and Dr Kong Hung Sze

Title: Phosphatidic acid phosphatase 1 (PAP-1) impairs SARS-CoV-2 replication by affecting the glycerophospholipid metabolism pathway.

Abstract: Viruses exploit the host lipid metabolism machinery to achieve efficient replication. We herein characterize the lipids profile reprogramming in vitro and in vivo using liquid chromatography-mass spectrometry-based untargeted lipidomics. The lipidome of SARS-CoV-2-infected Caco-2 cells was markedly different from that of mock-infected samples, with most of the changes involving downregulation of ceramides. In COVID-19 patients’ plasma samples, a total of 54 lipids belonging to 12 lipid classes that were significantly perturbed compared to non-infected control subjects’ plasma samples were identified. Among these 12 lipid classes, ether-linked phosphatidylcholines, ether-linked phosphatidylethanolamines, phosphatidylcholines, and ceramides were the four most perturbed. Pathway analysis revealed that the glycerophospholipid, sphingolipid, and ether lipid metabolisms pathway were the most significantly perturbed host pathways. Phosphatidic acid phosphatases (PAP) were involved in all three pathways and PAP-1 deficiency significantly suppressed SARS-CoV-2 replication. PAP-1 is encoded by a group of genes named LPINs and has three lipin proteins (lipin 1, 2 and 3), with each of them having PAP-1 activity. siRNA knockdown of LPIN2 and LPIN3 resulted in significant reduction of SARS-CoV-2 load in multiple cell lines. In summary, these findings characterized the host lipidomic changes upon SARS-CoV-2 infection and identified PAP-1 as a potential target for intervention for COVID-19.

 

Name: Dr Pui Wang

Title: An intranasal influenza virus-vectored vaccine protects against SARS-CoV-2 variants in mice and Syrian hamsters

Abstract: Vaccination is the most effective means of alleviating COVID-19 disease burden. However, emerging variants have led to continuing immune evasion since early 2021. Moreover, current mRNA vaccines do not elicit sufficient local mucosal immunity in the respiratory tract, which is important for preventing or reducing the transmission of SARS-CoV-2. There have been suggestions that the next generation of vaccines for SARS-CoV-2 should aim to offer higher protection by blocking infections altogether.

This study reports an influenza virus-vectored SARS-CoV-2 intranasal vaccine based on a live attenuated influenza virus with the NS1 gene deleted that expresses the receptor binding domain (RBD) of SARS-CoV-2 spike protein, designated as DelNS1-RBD4N-DAF.

Immune responses and protection against virus challenge after two doses of the BNT162b2 mRNA vaccine (intramuscular injection) or various DelNS1-RBD4N-DAF vaccines (intranasal) were analysed in mouse and hamster models. Our vaccines induced high levels of neutralizing antibodies against various variants, including Omicron BA.2 and stimulated robust T cell responses in mice. Intranasal inoculation with our vaccines elicited effective protection against challenge with SARS-CoV-2 variants in the upper and lower respiratory tracts of both animals.

In conclusion, our vaccine induced strong immune responses with cross protection against different SARS-CoV-2 variants. Our vaccine platform can be used to make bivalent vaccines against both influenza and COVID-19 in response to dual circulation of seasonal influenza and SARS-CoV-2 in the future.

Name: Dr Anna Jinxia Zhang

Title: COVID-19 mRNA vaccine restore impaired innate antiviral responses protects against SARS-CoV-2 Omicron infection in diet-induced obesity mice

Abstract: The pathogenicity of SARS-CoV-2 and host immune responses to infection and vaccination in individuals with obesity remain incompletely understood. We studied SARS-CoV-2 Alpha- and Omicron BA.1-induced disease and immune responses after infection or COVID-19 mRNA vaccination in diet-induced obese mice. Unlike in lean mice, Omicron BA.1 and Alpha replicated to comparable levels in the lungs of DIO mice and resulted in similar degree of tissue damages. Importantly, both T cell and B cell mediated adaptive immune responses to SARS-CoV-2 infection or COVID-19 mRNA vaccination are impaired in DIO mice, leading to higher propensity of re-infection and lower vaccine efficacy. However, in the absent of neutralizing antibody, vaccinated DIO mice are protected from lung damage upon Omicron challenge, accompanied with significant more IFN-α and IFN-β production in the lung tissue. Lung RNAseq and ex vivo culture of alveolar macrophages indicated COVID-19 mRNA vaccination in DIO mice boosted antiviral interferon response compared to nonvaccinated controls, with significant enhancement of IFN-α production from alveolar macrophages in responses to Poly(I:C) or recombinant SARS-CoV-2 spike protein stimulation. Our findings suggested that COVID-19 mRNA vaccination enhances host innate antiviral responses in obesity which is still beneficial to certain degree when adaptive immunity is suboptimal.

 

 

Name: Dr Hanjun Zhao

Title: A triple-functional peptide broadly inhibits SARS-CoV-2 variants in hamsters

Abstract: The emergence of highly transmissible SARS-CoV-2 variants has led to the waves of resurgence of COVID-19 cases. Effective antivirals against variants are required. Here we demonstrate that a human-derived peptide 4H30 has broad antiviral activity against the ancestral virus and four Variants of Concern (VOCs) in vitro. Mechanistically, 4H30 can inhibit three distinct steps of the SARS-CoV-2 life cycle. Specifically, 4H30 blocks viral entry by clustering SARS-CoV-2 virions; prevents membrane fusion by inhibiting endosomal acidification; and inhibits release of virions by cross-linking SARS-CoV-2 with cellular glycosaminoglycans. In vivo studies show that 4H30 significantly reduces the lung viral titers in hamsters, with a more potent reduction for Omicron variant than Delta variant. This is likely because the entry of Omicron variant mainly relies on the endocytic pathway which is targeted by 4H30. Moreover, 4H30 reduces syncytia formation in infected hamster lungs. These findings provide a proof of concept that a single antiviral can inhibit viral entry, fusion and release.

Organizer Information 

Acknowledgement

Health@InnoHK, Innovation and Technology Commission

Theme-based Research Scheme on Potentiating Host Immunity for HIV-1 Functional Cure

bottom of page