4.6 Article

Rethinking the Response to Emerging Microbes: Vaccines and Therapeutics in the Ebola Era-a Conference at Harvard Medical School

Journal

JOURNAL OF VIROLOGY
Volume 89, Issue 15, Pages 7446-7448

Publisher

AMER SOC MICROBIOLOGY
DOI: 10.1128/JVI.01251-15

Keywords

-

Categories

Funding

  1. Harvard Medical School Dean's Office and Department of Microbiology and Immunobiology

Ask authors/readers for more resources

Harvard Medical School convened a meeting of biomedical and clinical experts on 5 March 2015 on the topic of Rethinking the Response to Emerging Microbes: Vaccines and Therapeutics in the Ebola Era, with the goals of discussing the lessons from the recent Ebola outbreak and using those lessons as a case study to aid preparations for future emerging infections. The speakers and audience discussed the special challenges in combatting an infectious agent that causes sporadic outbreaks in resource-poor countries. The meeting led to a call for improved basic medical care for all and continued support of basic discovery research to provide the foundation for preparedness for future outbreaks in addition to the targeted emergency response to outbreaks and targeted research programs against Ebola virus and other specific emerging pathogens.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Microbiology

Complete Mapping of Mutations to the SARS-CoV-2 Spike Receptor-Binding Domain that Escape Antibody Recognition

Allison J. Greaney, Tyler N. Starr, Pavlo Gilchuk, Seth J. Zost, Elad Binshtein, Andrea N. Loes, Sarah K. Hilton, John Huddleston, Rachel Eguia, Katharine H. D. Crawford, Adam S. Dingens, Rachel S. Nargi, Rachel E. Sutton, Naveenchandra Suryadevara, Paul W. Rothlauf, Zhuoming Liu, Sean P. J. Whelan, Robert H. Carnahan, James E. Crowe, Jesse D. Bloom

Summary: Antibodies targeting the SARS-CoV-2 spike receptor-binding domain (RBD) are key in neutralizing antibody responses, and a deep mutational scanning method was used to assess the impact of all amino-acid mutations in the RBD on antibody binding with 10 human monoclonal antibodies. The study identified the clustered escape mutations in different surfaces of the RBD that correspond to structurally defined antibody epitopes, showing that even antibodies targeting the same surface can have distinct escape mutations.

CELL HOST & MICROBE (2021)

Article Immunology

SARS-CoV-2 Viral RNA Shedding for More Than 87 Days in an Individual With an Impaired CD8+T Cell Response

Jackson S. Turner, Aaron Day, Wafaa B. Alsoussi, Zhuoming Liu, Jane A. O'Halloran, Rachel M. Presti, Bruce K. Patterson, Sean P. J. Whelan, Ali H. Ellebedy, Philip A. Mudd

Summary: Prolonged shedding of viral RNA can occur in some individuals with COVID-19 for more than 3 months, and impaired CD8+ T cells may play a role in this process.

FRONTIERS IN IMMUNOLOGY (2021)

Article Biochemistry & Molecular Biology

N-terminal domain antigenic mapping reveals a site of vulnerability for SARS-CoV-2

Matthew McCallum, Anna De Marco, Florian Lempp, M. Alejandra Tortorici, Dora Pinto, Alexandra C. Walls, Martina Beltramello, Alex Chen, Zhuoming Liu, Fabrizia Zatta, Samantha Zepeda, Julia di Iulio, John E. Bowen, Martin Montiel-Ruiz, Jiayi Zhou, Laura E. Rosen, Siro Bianchi, Barbara Guarino, Chiara Silacci Fregni, Rana Abdelnabi, Shi-Yan Caroline Foo, Paul W. Rothlauf, Louis-Marie Bloyet, Fabio Benigni, Elisabetta Cameroni, Johan Neyts, Agostino Riva, Gyorgy Snell, Amalio Telenti, Sean P. J. Whelan, Herbert W. Virgin, Davide Corti, Matteo Samuele Pizzuto, David Veesler

Summary: The study identifies 41 human monoclonal antibodies that recognize the N-terminal domain of the SARS-CoV-2 spike protein and exhibit strong neutralizing activity. These antibodies inhibit cell-to-cell fusion, activate effector functions, and protect animals from virus challenge, highlighting the importance of NTD-specific neutralizing antibodies for protective immunity and vaccine development. Several SARS-CoV-2 variants with mutations in the NTD supersite suggest ongoing selective pressure on the virus.
Article Medicine, General & Internal

Effect of Immunosuppression on the Immunogenicity of mRNA Vaccines to SARS-CoV-2 A Prospective Cohort Study

Parakkal Deepak, Wooseob Kim, Michael A. Paley, Monica Yang, Alexander B. Carvidi, Emanuel G. Demissie, Alia A. El-Qunni, Alem Haile, Katherine Huang, Baylee Kinnett, Mariel J. Liebeskind, Zhuoming Liu, Lily E. McMorrow, Diana Paez, Niti Pawar, Dana C. Perantie, Rebecca E. Schriefer, Shannon E. Sides, Mahima Thapa, Mate Gergely, Suha Abushamma, Sewuese Akuse, Michael Klebert, Lynne Mitchell, Darren Nix, Jonathan Graf, Kimberly E. Taylor, Salim Chahin, Matthew A. Ciorba, Patricia Katz, Mehrdad Matloubian, Jane A. O'Halloran, Rachel M. Presti, Gregory F. Wu, Sean P. J. Whelan, William J. Buchser, Lianne S. Gensler, Mary C. Nakamura, Ali H. Ellebedy, Alfred H. J. Kim

Summary: Patients with CID show immune response to mRNA-based SARS-CoV-2 vaccines, but some may have lower antibody levels, especially those receiving glucocorticoids and B-cell depletion therapy. Further studies are needed to confirm these preliminary findings.

ANNALS OF INTERNAL MEDICINE (2021)

Article Biochemistry & Molecular Biology

Lrp1 is a host entry factor for Rift Valley fever virus

Safder S. Ganaie, Madeline M. Schwarz, Cynthia M. McMillen, David A. Price, Annie X. Feng, Joseph R. Albe, Wenjie Wang, Shane Miersch, Anthony Orvedahl, Aidan R. Cole, Monica F. Sentmanat, Nawneet Mishra, Devin A. Boyles, Zachary T. Koenig, Michael R. Kujawa, Matthew A. Demers, Ryan M. Hoehl, Austin B. Moyle, Nicole D. Wagner, Sarah H. Stubbs, Lia Cardarelli, Joan Teyra, Anita McElroy, Michael L. Gross, Sean P. J. Whelan, John Doench, Xiaoxia Cui, Tom J. Brett, Sachdev S. Sidhu, Herbert W. Virgin, Takeshi Egawa, Daisy W. Leung, Gaya K. Amarasinghe, Amy L. Hartman

Summary: RVFV infection depends on host factors such as Lrp1, Grp94, and RAP, which directly bind to the viral glycoprotein without glycosylation dependence. Exogenous addition of mRAP(D3) and anti-Lrp1 antibodies can neutralize RVFV infection. By regulating these factors, it is possible to limit the occurrence of RVFV infections.
Article Immunology

A vaccine-induced public antibody protects against SARS-CoV-2 and emerging variants

Aaron J. Schmitz, Jackson S. Turner, Zhuoming Liu, Julian Q. Zhou, Ishmael D. Aziati, Rita E. Chen, Astha Joshi, Traci L. Bricker, Tamarand L. Darling, Daniel C. Adelsberg, Clara G. Altomare, Wafaa B. Alsoussi, James Brett Case, Laura A. VanBlargan, Tingting Lei, Mahima Thapa, Fatima Amanat, Trushar Jeevan, Thomas Fabrizio, Jane A. O'Halloran, Pei-Yong Shi, Rachel M. Presti, Richard J. Webby, Florian Krammer, Sean P. J. Whelan, Goran Bajic, Michael S. Diamond, Adrianus C. M. Boon, Ali H. Ellebedy

Summary: The emergence of SARS-CoV-2 antigenic variants poses a public health threat, but analysis of monoclonal antibodies suggests that some antibodies can neutralize various variants. The study also found that an antibody called 2C08 has neutralizing effects against multiple variants and can reduce viral load and morbidity in animal models.

IMMUNITY (2021)

Article Multidisciplinary Sciences

In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains

Rita E. Chen, Emma S. Winkler, James Brett Case, Ishmael D. Aziati, Traci L. Bricker, Astha Joshi, Tamarand L. Darling, Baoling Ying, John M. Errico, Swathi Shrihari, Laura A. VanBlargan, Xuping Xie, Pavlo Gilchuk, Seth J. Zost, Lindsay Droit, Zhuoming Liu, Spencer Stumpf, David Wang, Scott A. Handley, W. Blaine Jr Jr Stine, Pei-Yong Shi, Meredith E. Davis-Gardner, Mehul S. Suthar, Miguel Garcia Knight, Raul Andino, Charles Y. Chiu, Ali H. Ellebedy, Daved H. Fremont, Sean P. J. Whelan, James E. Jr Jr Crowe, Lisa Purcell, Davide Corti, Adrianus C. M. Boon, Michael S. Diamond

Summary: Cell culture experiments showed reduced or abrogated neutralizing activity of monoclonal antibodies against SARS-CoV-2 variant strains, but low prophylactic doses of antibody combinations protected against infection in vivo without resistance emergence. Higher doses of several monoclonal antibody cocktails also provided protection against viruses with a B.1.351 spike gene in vivo. Many antibody products with Emergency Use Authorization should therefore retain substantial efficacy against prevailing variant strains of SARS-CoV-2.

NATURE (2021)

Article Multidisciplinary Sciences

Broad sarbecovirus neutralization by a human monoclonal antibody

M. Alejandra Tortorici, Nadine Czudnochowski, Tyler N. Starr, Roberta Marzi, Alexandra C. Walls, Fabrizia Zatta, John E. Bowen, Stefano Jaconi, Julia Di Iulio, Zhaoqian Wang, Anna De Marco, Samantha K. Zepeda, Dora Pinto, Zhuoming Liu, Martina Beltramello, Istvan Bartha, Michael P. Housley, Florian A. Lempp, Laura E. Rosen, Exequiel Dellota, Hannah Kaiser, Martin Montiel-Ruiz, Jiayi Zhou, Amin Addetia, Barbara Guarino, Katja Culap, Nicole Sprugasci, Christian Saliba, Eneida Vetti, Isabella Giacchetto-Sasselli, Chiara Silacci Fregni, Rana Abdelnabi, Shi-Yan Caroline Foo, Colin Havenar-Daughton, Michael A. Schmid, Fabio Benigni, Elisabetta Cameroni, Johan Neyts, Amalio Telenti, Herbert W. Virgin, Sean P. J. Whelan, Gyorgy Snell, Jesse D. Bloom, Davide Corti, David Veesler, Matteo Samuele Pizzuto

Summary: The emergence of SARS-CoV-2 variants and recurrent spillovers of coronaviruses into the human population emphasize the need for broadly neutralizing antibodies to prevent future zoonotic infections. The human monoclonal antibody S2X259 has shown promising results in neutralizing various forms of SARS-CoV-2 and potentially zoonotic sarbecoviruses by inhibiting the binding of ACE2 to the receptor-binding domain. This antibody targets a key antigenic site and may guide the design of vaccines effective against all sarbecoviruses.

NATURE (2021)

Article Multidisciplinary Sciences

Germinal centre-driven maturation of B cell response to mRNA vaccination

Wooseob Kim, Julian Q. Zhou, Stephen C. Horvath, Aaron J. Schmitz, Alexandria J. Sturtz, Tingting Lei, Zhuoming Liu, Elizaveta Kalaidina, Mahima Thapa, Wafaa B. Alsoussi, Alem Haile, Michael K. Klebert, Teresa Suessen, Luis Parra-Rodriguez, Philip A. Mudd, Sean P. J. Whelan, William D. Middleton, Sharlene A. Teefey, Iskra Pusic, Jane A. O'Halloran, Rachel M. Presti, Jackson S. Turner, Ali H. Ellebedy

Summary: SARS-CoV-2 mRNA vaccination induces a persistent germinal center reaction in humans, resulting in affinity-matured long-term antibody responses that potently neutralize the virus.

NATURE (2022)

Article Multidisciplinary Sciences

Antibody-mediated broad sarbecovirus neutralization through ACE2 molecular mimicry

Young-Jun Park, Anna De Marco, Tyler N. Starr, Zhuoming Liu, Dora Pinto, Alexandra C. Walls, Fabrizia Zatta, Samantha K. Zepeda, John Bowen, Kaitlin S. Sprouse, Anshu Joshi, Martina Giurdanella, Barbara Guarino, Julia Noack, Rana Abdelnabi, Shi-Yan Caroline Foo, Florian A. Lempp, Fabio Benigni, Gyorgy Snell, Johan Neyts, Sean Pj Whelan, Herbert W. Virgin, Jesse D. Bloom, Davide Corti, Matteo Samuele Pizzuto, David Veesler

Summary: Understanding broadly neutralizing sarbecovirus antibody responses is crucial for developing countermeasures against SARS-CoV-2 variants and future sarbecoviruses. The human monoclonal antibody S2K146 has been found to broadly neutralize ACE2-binding viruses and shows potential for clinical development. Conserved ACE2-binding residues could be leveraged for developing vaccines that elicit broad immunity.

SCIENCE (2022)

Article Multidisciplinary Sciences

Defining the risk of SARS-CoV-2 variants on immune protection

Marciela M. DeGrace, Elodie Ghedin, Matthew B. Frieman, Florian Krammer, Alba Grifoni, Arghavan Alisoltani, Galit Alter, Rama R. Amara, Ralph S. Baric, Dan H. Barouch, Jesse D. Bloom, Louis-Marie Bloyet, Gaston Bonenfant, Adrianus C. M. Boon, Eli A. Boritz, Debbie L. Bratt, Traci L. Bricker, Liliana Brown, William J. Buchser, Juan Manuel Carreno, Liel Cohen-Lavi, Tamarand L. Darling, Meredith E. Davis-Gardner, Bethany L. Dearlove, Han Di, Meike Dittmann, Nicole A. Doria-Rose, Daniel C. Douek, Christian Drosten, Venkata-Viswanadh Edara, Ali Ellebedy, Thomas P. Fabrizio, Guido Ferrari, Will M. Fischer, William C. Florence, Ron A. M. Fouchier, John Franks, Adolfo Garcia-Sastre, Adam Godzik, Ana Silvia Gonzalez-Reiche, Aubree Gordon, Bart L. Haagmans, Peter J. Halfmann, David D. Ho, Michael R. Holbrook, Yaoxing Huang, Sarah L. James, Lukasz Jaroszewski, Trushar Jeevan, Robert M. Johnson, Terry C. Jones, Astha Joshi, Yoshihiro Kawaoka, Lisa Kercher, Marion P. G. Koopmans, Bette Korber, Eilay Koren, Richard A. Koup, Eric B. LeGresley, Jacob E. Lemieux, Mariel J. Liebeskind, Zhuoming Liu, Brandi Livingston, James P. Logue, Yang Luo, Adrian B. McDermott, Margaret J. McElrath, Victoria A. Meliopoulos, Vineet D. Menachery, David C. Montefiori, Barbara Muehlemann, Vincent J. Munster, Jenny E. Munt, Manoj S. Nair, Antonia Netzl, Anna M. Niewiadomska, Sijy O'Dell, Andrew Pekosz, Stanley Perlman, Marjorie C. Pontelli, Barry Rockx, Morgane Rolland, Paul W. Rothlauf, Sinai Sacharen, Richard H. Scheuermann, Stephen D. Schmidt, Michael Schotsaert, Stacey Schultz-Cherry, Robert A. Seder, Mayya Sedova, Alessandro Sette, Reed S. Shabman, Xiaoying Shen, Pei-Yong Shi, Maulik Shukla, Viviana Simon, Spencer Stumpf, Nancy J. Sullivan, Larissa B. Thackray, James Theiler, Paul G. Thomas, Sanja Trifkovic, Sina Tureli, Samuel A. Turner, Maria A. Vakaki, Harm van Bakel, Laura A. VanBlargan, Leah R. Vincent, Zachary S. Wallace, Li Wang, Maple Wang, Pengfei Wang, Wei Wang, Scott C. Weaver, Richard J. Webby, Carol D. Weiss, David E. Wentworth, Stuart M. Weston, Sean P. J. Whelan, Bradley M. Whitener, Samuel H. Wilks, Xuping Xie, Baoling Ying, Hyejin Yoon, Bin Zhou, Tomer Hertz, Derek J. Smith, Michael S. Diamond, Diane J. Post, Mehul S. Suthar

Summary: The SAVE program is a real-time risk assessment initiative established by the National Institute of Allergy and Infectious Diseases to address the public health threat posed by the emergence of SARS-CoV-2 variants. Its goal is to evaluate the potential impact of these variants on transmission, virulence, and immunity induced through infection or vaccination.

NATURE (2022)

Article Multidisciplinary Sciences

CD164 is a host factor for lymphocytic choriomeningitis virus entry

Mark J. G. Bakkers, Alex Moon-Walker, Rasmus Herlo, Vesna Brusic, Sarah Hulsey Stubbs, Kathryn M. Hastie, Erica Ollmann Saphire, Tomas L. Kirchhausen, Sean P. J. Whelan

Summary: LCMV is a virus that enters cells by binding to different receptors and causes severe health problems. This study found that LCMV first binds to CD164 before entering cells, providing new targets for therapeutic interventions.

PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA (2022)

Article Multidisciplinary Sciences

Imprinted antibody responses against SARS-CoV-2 Omicron sublineages

Young-Jun Park, Dora Pinto, Alexandra C. Walls, Zhuoming Liu, Anna De Marco, Fabio Benigni, Fabrizia Zatta, Chiara Silacci-Fregni, Jessica Bassi, Kaitlin R. Sprouse, Amin Addetia, John E. Bowen, Cameron Stewart, Martina Giurdanella, Christian Saliba, Barbara Guarino, Michael A. Schmid, Nicholas M. Franko, Jennifer K. Logue, Ha V. Dang, Kevin Hauser, Julia di Iulio, William Rivera, Gretja Schnell, Anushka Rajesh, Jiayi Zhou, Nisar Farhat, Hannah Kaiser, Martin Montiel-Ruiz, Julia Noack, Florian A. Lempp, Javier Janer, Rana Abdelnabi, Piet Maes, Paolo Ferrari, Alessandro Ceschi, Olivier Giannini, Guilherme Dias De Melo, Lauriane Kergoat, Herve Bourhy, Johan Neyts, Leah Soriaga, Lisa A. Purcell, Gyorgy Snell, Sean P. J. Whelan, Antonio Lanzavecchia, Herbert W. Virgin, Luca Piccoli, Helen Y. Chu, Matteo Samuele Pizzuto, Davide Corti, David Veesler

Summary: SARS-CoV-2 Omicron sublineages have spike mutations that allow them to evade antibodies from previous infection or vaccination. Hybrid immunity or booster shots can generate neutralizing antibodies against Omicron variants, and breakthrough infections lead to the production of neutralizing antibodies in the nasal mucosa. Antibodies derived from memory B cells or plasma cells of Omicron breakthrough cases show cross-reactivity with different receptor-binding domains, while primary Omicron infections elicit B cells with narrow specificity. A highly potent pan-variant-neutralizing antibody has been identified as a potential candidate for clinical development.

SCIENCE (2022)

Article Biology

SARS-CoV-2 uses CD4 to infect T helper lymphocytes

Natalia S. Brunetti, Gustavo G. Davanzo, Diogo de Moraes, Allan J. R. Ferrari, Gabriela F. Souza, Stefanie Primon Muraro, Thiago L. Knittel, Vinicius O. Boldrini, Lauar B. Monteiro, Joo Victor Virgilio-da-Silva, Gerson S. Profeta, Natalia S. Wassano, Luana Nunes Santos, Victor C. Carregari, Artur H. S. Dias, Flavio P. Veras, Lucas A. Tavares, Julia Forato, Icaro M. S. Castro, Licia C. Silva-Costa, Andre C. Palma, Eli Mansour, Raisa G. Ulaf, Ana F. Bernardes, Thyago A. Nunes, Luciana C. Ribeiro, Marcus V. Agrela, Maria Luiza Moretti, Lucas I. Buscaratti, Fernanda Crunfli, Raissa G. Ludwig, Jaqueline A. Gerhardt, Natalia Munhoz-Alves, Ana Maria Marques, Renata Sesti-Costa, Mariene R. Amorim, Daniel A. Toledo-Teixeira, Pierina Lorencini Parise, Matheus Cavalheiro Martini, Karina Bispos-dos-Santos, Camila L. Simeoni, Fabiana Granja, Virginia C. Silvestrini, Eduardo B. de Oliveira, Vitor M. Faca, Murilo Carvalho, Bianca G. Castelucci, Alexandre B. Pereira, Lais D. Coimbra, Marieli M. G. Dias, Patricia B. Rodrigues, Arilson Bernardo S. P. Gomes, Fabricio B. Pereira, Leonilda M. B. Santos, Louis-Marie Bloyet, Spencer Stumpf, Marjorie C. Pontelli, Sean Whelan, Andrei C. Sposito, Robson F. Carvalho, Andre S. Vieira, Marco A. R. Vinolo, Andre Damasio, Licio Velloso, Ana Carolina M. Figueira, Luis L. P. da Silva, Thiago Mattar Cunha, Helder I. Nakaya, Henrique Marques-Souza, Rafael E. Marques, Daniel Martins-de-Souza, Munir S. Skaf, Jose Luiz Proenca-Modena, Pedro M. M. Moraes-Vieira, Marcelo A. Mori, Alessandro S. Farias

Summary: Research has found that SARS-CoV-2 mainly infects T helper cells (CD4+), but not T cells (CD8+). The infected T helper cells express higher levels of IL-10, which may contribute to viral persistence and increased disease severity, leading to poor immune response in COVID-19 patients.

ELIFE (2023)

Meeting Abstract Rheumatology

Additional Dose of SARS-CoV-2 Vaccine Improves Cross-Variant Neutralization Titers in Immunosuppressed Patients with Chronic Inflammatory Disease

Michael Paley, Parakkal Deepak, Wooseob Kim, Monica Yang, Vinay Chandrasekaran, Guadalupe Oliva Escudero, Katherine Huang, Zhuoming Liu, Lily McMorrow, Mahima Thapa, Matthew Ciorba, Mehrdad Matloubian, Lianne Gensler, Mary Nakamura, Sean Whelan, William Buchser, Ali Ellebedy, Alfred Kim

ARTHRITIS & RHEUMATOLOGY (2022)

No Data Available