Searched for: in-biosketch:true
person:mullim04
LY-CoV555, a rapidly isolated potent neutralizing antibody, provides protection in a non-human primate model of SARS-CoV-2 infection [PrePrint]
Jones, Bryan E; Brown-Augsburger, Patricia L; Corbett, Kizzmekia S; Westendorf, Kathryn; Davies, Julian; Cujec, Thomas P; Wiethoff, Christopher M; Blackbourne, Jamie L; Heinz, Beverly A; Foster, Denisa; Higgs, Richard E; Balasubramaniam, Deepa; Wang, Lingshu; Bidshahri, Roza; Kraft, Lucas; Hwang, Yuri; Žentelis, Stefanie; Jepson, Kevin R; Goya, Rodrigo; Smith, Maia A; Collins, David W; Hinshaw, Samuel J; Tycho, Sean A; Pellacani, Davide; Xiang, Ping; Muthuraman, Krithika; Sobhanifar, Solmaz; Piper, Marissa H; Triana, Franz J; Hendle, Jorg; Pustilnik, Anna; Adams, Andrew C; Berens, Shawn J; Baric, Ralph S; Martinez, David R; Cross, Robert W; Geisbert, Thomas W; Borisevich, Viktoriya; Abiona, Olubukola; Belli, Hayley M; de Vries, Maren; Mohamed, Adil; Dittmann, Meike; Samanovic, Marie; Mulligan, Mark J; Goldsmith, Jory A; Hsieh, Ching-Lin; Johnson, Nicole V; Wrapp, Daniel; McLellan, Jason S; Barnhart, Bryan C; Graham, Barney S; Mascola, John R; Hansen, Carl L; Falconer, Ester
SARS-CoV-2 poses a public health threat for which therapeutic agents are urgently needed. Herein, we report that high-throughput microfluidic screening of antigen-specific B-cells led to the identification of LY-CoV555, a potent anti-spike neutralizing antibody from a convalescent COVID-19 patient. Biochemical, structural, and functional characterization revealed high-affinity binding to the receptor-binding domain, ACE2 binding inhibition, and potent neutralizing activity. In a rhesus macaque challenge model, prophylaxis doses as low as 2.5 mg/kg reduced viral replication in the upper and lower respiratory tract. These data demonstrate that high-throughput screening can lead to the identification of a potent antiviral antibody that protects against SARS-CoV-2 infection.
PMCID:7536866
PMID: 33024963
ISSN: 2692-8205
CID: 4626872
Treating COVID-19 With Hydroxychloroquine (TEACH): A Multicenter, Double-Blind Randomized Controlled Trial in Hospitalized Patients
Ulrich, Robert J; Troxel, Andrea B; Carmody, Ellie; Eapen, Jaishvi; Bäcker, Martin; DeHovitz, Jack A; Prasad, Prithiv J; Li, Yi; Delgado, Camila; Jrada, Morris; Robbins, Gabriel A; Henderson, Brooklyn; Hrycko, Alexander; Delpachitra, Dinuli; Raabe, Vanessa; Austrian, Jonathan S; Dubrovskaya, Yanina; Mulligan, Mark J
Background/UNASSIGNED:Effective therapies to combat coronavirus 2019 (COVID-19) are urgently needed. Hydroxychloroquine (HCQ) has in vitro antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the clinical benefit of HCQ in treating COVID-19 is unclear. Randomized controlled trials are needed to determine the safety and efficacy of HCQ for the treatment of hospitalized patients with COVID-19. Methods/UNASSIGNED:We conducted a multicenter, double-blind randomized clinical trial of HCQ among patients hospitalized with laboratory-confirmed COVID-19. Subjects were randomized in a 1:1 ratio to HCQ or placebo for 5 days and followed for 30 days. The primary efficacy outcome was a severe disease progression composite end point (death, intensive care unit admission, mechanical ventilation, extracorporeal membrane oxygenation, and/or vasopressor use) at day 14. Results/UNASSIGNED: = .350). There were no significant differences in COVID-19 clinical scores, number of oxygen-free days, SARS-CoV-2 clearance, or adverse events between HCQ and placebo. HCQ was associated with a slight increase in mean corrected QT interval, an increased D-dimer, and a trend toward an increased length of stay. Conclusions/UNASSIGNED:In hospitalized patients with COVID-19, our data suggest that HCQ does not prevent severe outcomes or improve clinical scores. However, our conclusions are limited by a relatively small sample size, and larger randomized controlled trials or pooled analyses are needed.
PMCID:7543602
PMID: 33134417
ISSN: 2328-8957
CID: 4655862
Phase 1/2 study of COVID-19 RNA vaccine BNT162b1 in adults
Mulligan, Mark J; Lyke, Kirsten E; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Neuzil, Kathleen; Raabe, Vanessa; Bailey, Ruth; Swanson, Kena A; Li, Ping; Koury, Kenneth; Kalina, Warren; Cooper, David; Fontes-Garfias, Camila; Shi, Pei-Yong; Türeci, Özlem; Tompkins, Kristin R; Walsh, Edward E; Frenck, Robert; Falsey, Ann R; Dormitzer, Philip R; Gruber, William C; Şahin, Uğur; Jansen, Kathrin U
In March 2020, the World Health Organization (WHO) declared a pandemic of coronavirus disease 2019 (COVID-19), due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)1. With rapidly accumulating cases and deaths reported globally2, a vaccine is urgently needed. We report the available safety, tolerability, and immunogenicity data from an ongoing placebo-controlled, observer-blinded dose escalation study among 45 healthy adults, 18 to 55 years of age, randomized to receive 2 doses, separated by 21 days, of 10 µg, 30 µg, or 100 µg of BNT162b1, a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccine that encodes trimerized SARS-CoV-2 spike glycoprotein receptor-binding domain (RBD). Local reactions and systemic events were dose-dependent, generally mild to moderate, and transient. A second vaccination with 100 µg was not administered due to increased reactogenicity and a lack of meaningfully increased immunogenicity after a single dose compared to the 30 μg dose. RBD-binding IgG concentrations and SARS-CoV-2 neutralizing titers in sera increased with dose level and after a second dose. Geometric mean neutralizing titers reached 1.9- to 4.6-fold that of a panel of COVID-19 convalescent human sera at least 14 days after a positive SARS-CoV-2 PCR. These results support further evaluation of this mRNA vaccine candidate. (ClinicalTrials.gov identifier: NCT04368728).
PMID: 32785213
ISSN: 1476-4687
CID: 4555102
An Inactivated Virus Candidate Vaccine to Prevent COVID-19
Mulligan, Mark J
PMID: 32789500
ISSN: 1538-3598
CID: 4555112
RNA-Based COVID-19 Vaccine BNT162b2 Selected for a Pivotal Efficacy Study
Walsh, Edward E; Frenck, Robert; Falsey, Ann R; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Neuzil, Kathleen; Mulligan, Mark J; Bailey, Ruth; Swanson, Kena A; Li, Ping; Koury, Kenneth; Kalina, Warren; Cooper, David; Fontes-Garfias, Camila; Shi, Pei-Yong; Türeci, Özlem; Thompkins, Kristin R; Lyke, Kirsten E; Raabe, Vanessa; Dormitzer, Philip R; Jansen, Kathrin U; Sahin, Uğur; Gruber, William C
BACKGROUND:Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and the resulting disease, coronavirus disease 2019 (COVID-19), have spread to millions of people globally. Multiple vaccine candidates are under development, but no vaccine is currently available. METHODS:Healthy adults 18-55 and 65-85 years of age were randomized in an ongoing, placebo-controlled, observer-blinded dose-escalation study to receive 2 doses at 21-day intervals of placebo or either of 2 lipid nanoparticle-formulated, nucleoside-modified RNA vaccine candidates: BNT162b1, which encodes a secreted trimerized SARS-CoV-2 receptor-binding domain, or BNT162b2, which encodes a prefusion stabilized membrane-anchored SARS-CoV-2 full-length spike. In each of 13 groups of 15 participants, 12 received vaccine and 3 received placebo. Groups were distinguished by vaccine candidate, age of participant, and vaccine dose level. Interim safety and immunogenicity data of BNT162b1 in younger adults have been reported previously from US and German trials. We now present additional safety and immunogenicity data from the US Phase 1 trial that supported selection of the vaccine candidate advanced to a pivotal Phase 2/3 safety and efficacy evaluation. RESULTS:In both younger and older adults, the 2 vaccine candidates elicited similar dose-dependent SARS-CoV-2-neutralizing geometric mean titers (GMTs), comparable to or higher than the GMT of a panel of SARS-CoV-2 convalescent sera. BNT162b2 was associated with less systemic reactogenicity, particularly in older adults. CONCLUSION/CONCLUSIONS:These results support selection of the BNT162b2 vaccine candidate for Phase 2/3 large-scale safety and efficacy evaluation, currently underway.
PMCID:7444302
PMID: 32839784
ISSN: n/a
CID: 4575412
Sequencing identifies multiple, early introductions of SARS-CoV2 to New York City Region
Maurano, Matthew T; Ramaswami, Sitharam; Westby, Gael; Zappile, Paul; Dimartino, Dacia; Shen, Guomiao; Feng, Xiaojun; Ribeiro-Dos-Santos, Andre M; Vulpescu, Nicholas A; Black, Margaret; Hogan, Megan; Marier, Christian; Meyn, Peter; Zhang, Yutong; Cadley, John; Ordonez, Raquel; Luther, Raven; Huang, Emily; Guzman, Emily; Serrano, Antonio; Belovarac, Brendan; Gindin, Tatyana; Lytle, Andrew; Pinnell, Jared; Vougiouklakis, Theodore; Boytard, Ludovic; Chen, John; Lin, Lawrence H; Rapkiewicz, Amy; Raabe, Vanessa; Samanovic-Golden, Marie I; Jour, George; Osman, Iman; Aguero-Rosenfeld, Maria; Mulligan, Mark J; Cotzia, Paolo; Snuderl, Matija; Heguy, Adriana
Effective public response to a pandemic relies upon accurate measurement of the extent and dynamics of an outbreak. Viral genome sequencing has emerged as a powerful approach to link seemingly unrelated cases, and large-scale sequencing surveillance can inform on critical epidemiological parameters. Here, we report the analysis of 236 SARS-CoV2 sequences from cases in the New York City metropolitan area during the initial stages of the 2020 COVID-19 outbreak. The majority of cases throughout the region had no recent travel history or known exposure, and genetically linked cases were spread throughout the region. Comparison to global viral sequences showed that the majority were most related to cases from Europe. Our data are consistent with numerous seed transmissions from multiple sources and a prolonged period of unrecognized community spreading. This work highlights the complementary role of real-time genomic surveillance in addition to traditional epidemiological indicators.
PMCID:7276014
PMID: 32511587
ISSN: n/a
CID: 4477902
Adjuvanted H5N1 influenza vaccine enhances both cross-reactive memory B cell and strain-specific naive B cell responses in humans
Ellebedy, Ali H; Nachbagauer, Raffael; Jackson, Katherine J L; Dai, Ya-Nan; Han, Julianna; Alsoussi, Wafaa B; Davis, Carl W; Stadlbauer, Daniel; Rouphael, Nadine; Chromikova, Veronika; McCausland, Megan; Chang, Cathy Y; Cortese, Mario; Bower, Mary; Chennareddy, Chakravarthy; Schmitz, Aaron J; Zarnitsyna, Veronika I; Lai, Lilin; Rajabhathor, Arvind; Kazemian, Cheyann; Antia, Rustom; Mulligan, Mark J; Ward, Andrew B; Fremont, Daved H; Boyd, Scott D; Pulendran, Bali; Krammer, Florian; Ahmed, Rafi
There is a need for improved influenza vaccines. In this study we compared the antibody responses in humans after vaccination with an AS03-adjuvanted versus nonadjuvanted H5N1 avian influenza virus inactivated vaccine. Healthy young adults received two doses of either formulation 3 wk apart. We found that AS03 significantly enhanced H5 hemagglutinin (HA)-specific plasmablast and antibody responses compared to the nonadjuvanted vaccine. Plasmablast response after the first immunization was exclusively directed to the conserved HA stem region and came from memory B cells. Monoclonal antibodies (mAbs) derived from these plasmablasts had high levels of somatic hypermutation (SHM) and recognized the HA stem region of multiple influenza virus subtypes. Second immunization induced a plasmablast response to the highly variable HA head region. mAbs derived from these plasmablasts exhibited minimal SHM (naive B cell origin) and largely recognized the HA head region of the immunizing H5N1 strain. Interestingly, the antibody response to H5 HA stem region was much lower after the second immunization, and this suppression was most likely due to blocking of these epitopes by stem-specific antibodies induced by the first immunization. Taken together, these findings show that an adjuvanted influenza vaccine can substantially increase antibody responses in humans by effectively recruiting preexisting memory B cells as well as naive B cells into the response. In addition, we show that high levels of preexisting antibody can have a negative effect on boosting. These findings have implications toward the development of a universal influenza vaccine.
PMID: 32661157
ISSN: 1091-6490
CID: 4528002
Transcriptomic and Metabolic Responses to a Live-Attenuated Francisella tularensis Vaccine
Goll, Johannes B; Li, Shuzhao; Edwards, James L; Bosinger, Steven E; Jensen, Travis L; Wang, Yating; Hooper, William F; Gelber, Casey E; Sanders, Katherine L; Anderson, Evan J; Rouphael, Nadine; Natrajan, Muktha S; Johnson, Robert A; Sanz, Patrick; Hoft, Daniel; Mulligan, Mark J
The immune response to live-attenuated Francisella tularensis vaccine and its host evasion mechanisms are incompletely understood. Using RNA-Seq and LC-MS on samples collected pre-vaccination and at days 1, 2, 7, and 14 post-vaccination, we identified differentially expressed genes in PBMCs, metabolites in serum, enriched pathways, and metabolites that correlated with T cell and B cell responses, or gene expression modules. While an early activation of interferon α/β signaling was observed, several innate immune signaling pathways including TLR, TNF, NF-κB, and NOD-like receptor signaling and key inflammatory cytokines such as Il-1α, Il-1β, and TNF typically activated following infection were suppressed. The NF-κB pathway was the most impacted and the likely route of attack. Plasma cells, immunoglobulin, and B cell signatures were evident by day 7. MHC I antigen presentation was more actively up-regulated first followed by MHC II which coincided with the emergence of humoral immune signatures. Metabolomics analysis showed that glycolysis and TCA cycle-related metabolites were perturbed including a decline in pyruvate. Correlation networks that provide hypotheses on the interplay between changes in innate immune, T cell, and B cell gene expression signatures and metabolites are provided. Results demonstrate the utility of transcriptomics and metabolomics for better understanding molecular mechanisms of vaccine response and potential host-pathogen interactions.
PMID: 32722194
ISSN: 2076-393x
CID: 4541112
Alterations in the Human Plasma Lipidome in Response to Tularemia Vaccination
Maner-Smith, Kristal M; Goll, Johannes B; Khadka, Manoj; Jensen, Travis L; Colucci, Jennifer K; Gelber, Casey E; Albert, Carolyn J; Bosinger, Steven E; Franke, Jacob D; Natrajan, Muktha; Rouphael, Nadine; Johnson, Robert A; Sanz, Patrick; Anderson, Evan J; Hoft, Daniel F; Mulligan, Mark J; Ford, David A; Ortlund, Eric A
Tularemia is a highly infectious and contagious disease caused by the bacterium Francisella tularensis. To better understand human response to a live-attenuated tularemia vaccine and the biological pathways altered post-vaccination, healthy adults were vaccinated, and plasma was collected pre- and post-vaccination for longitudinal lipidomics studies. Using tandem mass spectrometry, we fully characterized individual lipid species within predominant lipid classes to identify changes in the plasma lipidome during the vaccine response. Separately, we targeted oxylipins, a subset of lipid mediators involved in inflammatory pathways. We identified 14 differentially abundant lipid species from eight lipid classes. These included 5-hydroxyeicosatetraenoic acid (5-HETE) which is indicative of lipoxygenase activity and, subsequently, inflammation. Results suggest that 5-HETE was metabolized to a dihydroxyeicosatrienoic acid (DHET) by day 7 post-vaccination, shedding light on the kinetics of the 5-HETE-mediated inflammatory response. In addition to 5-HETE and DHET, we observed pronounced changes in 34:1 phosphatidylinositol, anandamide, oleamide, ceramides, 16:1 cholesteryl ester, and other glycerophospholipids; several of these changes in abundance were correlated with serum cytokines and T cell activation. These data provide new insights into alterations in plasma lipidome post-tularemia vaccination, potentially identifying key mediators and pathways involved in vaccine response and efficacy.
PMID: 32722213
ISSN: 2076-393x
CID: 4541132
Proteomic Analysis of Human Immune Responses to Live-Attenuated Tularemia Vaccine
Chang, Yie-Hwa; Duong, Duc M; Goll, Johannes B; Wood, David C; Jensen, Travis L; Yin, Luming; Gelber, Casey E; Seyfried, Nicholas T; Anderson, Evan; Natrajan, Muktha S; Rouphael, Nadine; Johnson, Robert A; Sanz, Patrick; Mulligan, Mark J; Hoft, Daniel F
Francisella tularensis (F. tularensis) is an intracellular pathogen that causes a potentially debilitating febrile illness known as tularemia. F. tularensis can be spread by aerosol transmission and cause fatal pneumonic tularemia. If untreated, mortality rates can be as high as 30%. To study the host responses to a live-attenuated tularemia vaccine, peripheral blood mononuclear cell (PBMC) samples were assayed from 10 subjects collected pre- and post-vaccination, using both the 2D-DIGE/MALDI-MS/MS and LC-MS/MS approaches. Protein expression related to antigen processing and presentation, inflammation (PPARγ nuclear receptor), phagocytosis, and gram-negative bacterial infection was enriched at Day 7 and/or Day 14. Protein candidates that could be used to predict human immune responses were identified by evaluating the correlation between proteome changes and humoral and cellular immune responses. Consistent with the proteomics data, parallel transcriptomics data showed that MHC class I and class II-related signals important for protein processing and antigen presentation were up-regulated, further confirming the proteomic results. These findings provide new biological insights that can be built upon in future clinical studies, using live attenuated strains as immunogens, including their potential use as surrogates of protection.
PMID: 32722207
ISSN: 2076-393x
CID: 4541122