William W. Tang
La Jolla Institute for Allergy and Immunology
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by William W. Tang.
Cell | 2017
Justin M. Richner; Sunny Himansu; Kimberly A. Dowd; Scott L. Butler; Vanessa Salazar; Julie M. Fox; Justin G. Julander; William W. Tang; Sujan Shresta; Theodore C. Pierson; Giuseppe Ciaramella; Michael S. Diamond
The emergence of ZIKV infection has prompted a global effort to develop safe and effective vaccines. We engineered a lipid nanoparticle (LNP) encapsulated modified mRNA vaccine encoding wild-type or variant ZIKV structural genes and tested immunogenicity and protection in mice. Two doses of modified mRNA LNPs encoding prM-E genes that produced virus-like particles resulted in high neutralizing antibody titers (∼1/100,000) that protected against ZIKV infection and conferred sterilizing immunity. To offset a theoretical concern of ZIKV vaccines inducing antibodies that cross-react with the related dengue virus (DENV), we designed modified prM-E RNA encoding mutations destroying the conserved fusion-loop epitope in the E protein. This variant protected against ZIKV and diminished production of antibodies enhancing DENV infection in cells or mice. A modified mRNA vaccine can prevent ZIKV disease and be adapted to reduce the risk of sensitizing individuals to subsequent exposure to DENV, should this become a clinically relevant concern.
Nature microbiology | 2017
Jinsheng Wen; William W. Tang; Nicholas Sheets; Julia Ellison; Alessandro Sette; Kenneth Kim; Sujan Shresta
CD8+ T cells play an important role in controlling Flavivirus infection, including Zika virus (ZIKV). Here, we have identified 25 HLA-B*0702-restricted epitopes and 1 HLA-A*0101-restricted epitope using interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) and intracellular cytokine staining (ICS) in ZIKV-infected IFN-α/β receptor-deficient HLA transgenic mice. The cross-reactivity of ZIKV epitopes to dengue virus (DENV) was tested using IFN-γ-ELISPOT and IFN-γ-ICS on CD8+ T cells from DENV-infected mice, and five cross-reactive HLA-B*0702-binding peptides were identified by both assays. ZIKV/DENV cross-reactive CD8+ T cells in DENV-immune mice expanded post ZIKV challenge and dominated in the subsequent CD8+ T cell response. ZIKV challenge following immunization of mice with ZIKV-specific and ZIKV/DENV cross-reactive epitopes elicited CD8+ T cell responses that reduced infectious ZIKV levels, and CD8+ T cell depletions confirmed that CD8+ T cells mediated this protection. These results identify ZIKV-specific and ZIKV/DENV cross-reactive epitopes and demonstrate both an altered immunodominance pattern in the DENV-immune setting relative to naive, as well as a protective role for epitope-specific CD8+ T cells against ZIKV. These results have important implications for ZIKV vaccine development and provide a mouse model for evaluating anti-ZIKV CD8+ T cell responses of human relevance.
Journal of Immunology | 2014
Raphaël M. Zellweger; William E. Eddy; William W. Tang; Robyn Miller; Sujan Shresta
Dengue virus (DENV) causes pathologies ranging from the febrile illness dengue fever to the potentially lethal severe dengue disease. A major risk factor for developing severe dengue disease is the presence of subprotective DENV-reactive Abs from a previous infection (or from an immune mother), which can induce Ab-dependent enhancement of infection (ADE). However, infection in the presence of subprotective anti–DENV Abs does not always result in severe disease, suggesting that other factors influence disease severity. In this study we investigated how CD8+ T cell responses influence the outcome of Ab-mediated severe dengue disease. Mice were primed with aluminum hydroxide–adjuvanted UV-inactivated DENV prior to challenge with DENV. Priming failed to induce robust CD8+ T cell responses, and it induced nonneutralizing Ab responses that increased disease severity upon infection. Transfer of exogenous DENV-activated CD8+ T cells into primed mice prior to infection prevented Ab-dependent enhancement and dramatically reduced viral load. Our results suggest that in the presence of subprotective anti–DENV Abs, efficient CD8+ T cell responses reduce the risk of Ab-mediated severe dengue disease.
Journal of Virology | 2015
Raphaël M. Zellweger; William W. Tang; William E. Eddy; Kevin King; Marisa Sanchez; Sujan Shresta
ABSTRACT Dengue virus (DENV) is a major public health threat worldwide. Infection with one of the four serotypes of DENV results in a transient period of protection against reinfection with all serotypes (cross-protection), followed by lifelong immunity to the infecting serotype. While a protective role for neutralizing antibody responses is well established, the contribution of T cells to reinfection is less clear, especially during heterotypic reinfection. This study investigates the role of T cells during homotypic and heterotypic DENV reinfection. Mice were sequentially infected with homotypic or heterotypic DENV serotypes, and T cell subsets were depleted before the second infection to assess the role of DENV-primed T cells during reinfection. Mice primed nonlethally with DENV were protected against reinfection with either a homotypic or heterotypic serotype 2 weeks later. Homotypic priming induced a robust neutralizing antibody response, whereas heterotypic priming elicited binding, but nonneutralizing antibodies. CD8+ T cells were required for protection against heterotypic, but not homotypic, reinfection. These results suggest that T cells can contribute crucially to protection against heterotypic reinfection in situations where humoral responses alone may not be protective. Our findings have important implications for vaccine design, as they suggest that inducing both humoral and cellular responses during vaccination may maximize protective efficacy across all DENV serotypes. IMPORTANCE Dengue virus is present in more than 120 countries in tropical and subtropical regions. Infection with dengue virus can be asymptomatic, but it can also progress into the potentially lethal severe dengue disease. There are four closely related dengue virus serotypes. Infection with one serotype results in a transient period of resistance against all serotypes (cross-protection), followed by lifelong resistance to the infecting serotype, but not the other ones. The duration and mechanisms of the transient cross-protection period remain elusive. This study investigates the contribution of cellular immunity to cross-protection using mouse models of DENV infection. Our results demonstrate that cellular immunity is crucial to mediate cross-protection against reinfection with a different serotype, but not for protection against reinfection with the same serotype. A better understanding of the mediators responsible for the cross-protection period is important for vaccine design, as an ideal vaccine against dengue virus should efficiently protect against all serotypes.
EBioMedicine | 2016
Annie Elong Ngono; Hui-Wen Chen; William W. Tang; Yunichel Joo; Kevin King; Daniela Weiskopf; John Sidney; Alessandro Sette; Sujan Shresta
Infection with one of the four dengue virus serotypes (DENV1-4) presumably leads to lifelong immunity against the infecting serotype but not against heterotypic reinfection, resulting in a greater risk of developing Dengue Hemorrhagic Fever/Dengue Shock Syndrome (DHF/DSS) during secondary infection. Both antibodies and T cell responses have been implicated in DHF/DSS pathogenesis. According to the T cell-based hypothesis termed “original antigenic sin,” secondary DENV infection is dominated by non-protective, cross-reactive T cells that elicit an aberrant immune response. The goal of our study was to compare the roles of serotype-specific and cross-reactive T cells in protection vs. pathogenesis during DENV infection in vivo. Specifically, we utilized IFN-α/βR−/− HLA*B0702 transgenic mice in the context of peptide vaccination with relevant human CD8 T cell epitopes. IFN-α/βR−/− HLA*B0702 transgenic mice were immunized with DENV serotype 2 (DENV2)-specific epitopes or variants found in any of the other three serotypes (DENV1, DENV3 or DENV4), followed by challenge with DENV. Although cross-reactive T cell responses were lower than responses elicited by serotype-specific T cells, immunization with either serotype-specific or variant peptide epitopes enhanced viral clearance, demonstrating that both serotype-specific and cross-reactive T cells can contribute to protection in vivo against DENV infection.
Antiviral Research | 2016
Kelly L. Warfield; Emily M. Plummer; Andrew C. Sayce; Dominic S. Alonzi; William W. Tang; Beatrice E. Tyrrell; Michelle L. Hill; Alessandro T. Caputo; Sarah S. Killingbeck; P. Robert Beatty; Eva Harris; Ren Iwaki; Kyoko Kinami; Daisuke Ide; J. L. Kiappes; Atsushi Kato; Michael D. Buck; Kevin King; William E. Eddy; Mansoora Khaliq; Aruna Sampath; Anthony M. Treston; Raymond A. Dwek; Sven Enterlein; Joanna L. Miller; Nicole Zitzmann; Urban Ramstedt; Sujan Shresta
The antiviral activity of UV-4 was previously demonstrated against dengue virus serotype 2 (DENV2) in multiple mouse models. Herein, step-wise minimal effective dose and therapeutic window of efficacy studies of UV-4B (UV-4 hydrochloride salt) were conducted in an antibody-dependent enhancement (ADE) mouse model of severe DENV2 infection in AG129 mice lacking types I and II interferon receptors. Significant survival benefit was demonstrated with 10–20 mg/kg of UV-4B administered thrice daily (TID) for seven days with initiation of treatment up to 48 h after infection. UV-4B also reduced infectious virus production in in vitro antiviral activity assays against all four DENV serotypes, including clinical isolates. A set of purified enzyme, in vitro, and in vivo studies demonstrated that inhibition of endoplasmic reticulum (ER) α-glucosidases and not the glycosphingolipid pathway appears to be responsible for the antiviral activity of UV-4B against DENV. Along with a comprehensive safety package, these and previously published data provided support for an Investigational New Drug (IND) filing and Phases 1 and 2 clinical trials for UV-4B with an indication of acute dengue disease.
Current Opinion in Virology | 2015
William W. Tang; Rajvir Grewal; Sujan Shresta
Dengue virus (DENV) is a globally important mosquito-borne virus that causes a spectrum of diseases ranging from dengue fever (DF) to dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS), affecting 3.6 billion people in 128 countries [1,2(•)]. There is an urgent need for a drug or vaccine against DENV, yet none are presently available. In fact, results from recent Phase IIb and III trials of an attenuated tetrameric vaccine revealed that the vaccine provided limited protection against DENV serotype 2 in DENV-immune people, and no protection against any serotype in naïve individuals [3-5], highlighting the difficulties associated with dengue vaccine development. A challenge in the development of a DENV vaccine is that a vaccine must protect against all four DENV serotypes, which co-circulate in endemic areas. Further complicating DENV vaccine development is that the correlates of protection are not fully defined, mechanisms regulating the generation of protective antibody and T cell responses against all four DENV serotypes are as yet to be deciphered, and the adaptive immune response may actually contribute to severe disease. Recent studies using the only available animal model of DHF/DSS in mice lacking one or more components of the interferon (IFN) system have begun to provide crucial insights into the protective versus pathogenic nature of both antibody and T cell responses to DENV. Herein, we highlight key studies using the IFN receptor-deficient mouse models toward understanding the contribution of antibodies and T cells in impacting the outcome of DENV infection.
Cell Reports | 2017
Aaron F. Carlin; Emily M. Plummer; Edward A. Vizcarra; Nicholas Sheets; Yunichel Joo; William W. Tang; Jeremy Day; Jay Greenbaum; Christopher K. Glass; Michael S. Diamond; Sujan Shresta
Interferon-regulatory factors (IRFs) are a family of transcription factors (TFs) that translate viral recognition into antiviral responses, including type I interferon (IFN) production. Dengue virus (DENV) and other clinically important flaviviruses are suppressed by type I IFN. While mice lacking the type I IFN receptor (Ifnar1-/-) succumb to DENV infection, we found that mice deficient in three transcription factors controlling type I IFN production (Irf3-/-Irf5-/-Irf7-/- triple knockout [TKO]) survive DENV challenge. DENV infection of TKO mice resulted in minimal type I IFN production but a robust type II IFN (IFN-γ) response. Using loss-of-function approaches for various molecules, we demonstrate that the IRF-3-, IRF-5-, IRF-7-independent pathway predominantly utilizes IFN-γ and, to a lesser degree, type I IFNs. This pathway signals via IRF-1 to stimulate interleukin-12 (IL-12) production and IFN-γ response. These results reveal a key antiviral role for IRF-1 by activating both type I and II IFN responses during DENV infection.
Antiviral Research | 2018
Emilie Branche; William W. Tang; Karla M. Viramontes; Matthew Perry Young; Nicholas Sheets; Yunichel Joo; Anh-Viet T. Nguyen; Sujan Shresta
Abstract Dengue virus (DENV) currently circulates in more than 100 countries and causes an estimated 390 million infections per year. While most cases manifest as a self‐resolving fever, ˜1.5% of infections develop into a more severe dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS), which causes ˜20,000 deaths annually. The underlying pathological feature of DHF/DSS, also known as Severe Dengue, is an acute increase in vascular permeability leading to hypovolemia and shock. Angiogenic factors and cytokines, such as vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF), have been implicated in the increased vascular permeability, suggesting a potential therapeutic strategy for Severe Dengue. Here, we employed a mouse model of antibody‐dependent enhancement of DENV infection, which recapitulates the fatal capillary leakage and shock of human Severe Dengue, to investigate the effects of approved VEGF‐ and TNF‐targeting drugs. DENV infection caused a significant increase in serum VEGF levels within 2 days and resulted in ˜80% mortality within 8 days of infection. Treatment of mice with sunitinib, a VEGF receptor tyrosine kinase inhibitor, once (day 2) or twice (days 1 and 2) post‐infection reduced mortality by 50–80% compared with untreated mice. Notably, sunitinib treatment decreased serum TNF levels, white blood cell counts, and hematocrit levels relative to untreated mice, but had only marginal effects on tissue viral burden. Combination therapy with anti‐TNF antibody and sunitinib significantly reduced vascular leakage and synergized to provide superior protection from lethal DENV infection compared with either agent alone. These data suggest that a two‐pronged anti‐angiogenic and anti‐inflammatory approach may be useful for the rapid treatment of DHF/DSS. HighlightsDual treatment with sunitinib and anti‐TNF prevents severe dengue in mice.Sunitinib and anti‐TNF synergize to reduce vascular leakage in mice.Dual treatment with sunitinib and anti‐TNF does not impact tissue viral burden in mice.
Cell Stem Cell | 2016
Hongda Li; Laura Saucedo-Cuevas; Jose Angel Regla-Nava; Guoliang Chai; Nicholas Sheets; William W. Tang; Alexey Terskikh; Sujan Shresta; Joseph G. Gleeson