Buscar Close Search
Page Menu

Faculty

  • Robert Finberg

    Robert Finberg, MD

    Category: Clinical Trials,Host Response

    The Richard M. Haidack Professor in Medicine and chair and professor of medicine

    Dr. Finberg oversees three clinical trials currently enrolling patients with COVID-19. Two are of the drug remdesivir in ICU patients: one is an NIH-funded multicenter controlled trial and the other is an expanded access trial funded by the manufacturer Gilead.

    The third trial is a Phase II clinical trial to evaluate the safety and efficacy of the protease inhibitor favipiravir, funded by the manufacturer Fujifilm and in collaboration with Brigham and Women’s Hospital and Massachusetts General Hospital. 

    Four proposed clinical trials are being reviewed.

    Dr. Finberg, along with Scot Wolfe, PhD, has ongoing studies using CRISPR/Cas9 gene editing tools to define the virus and host genetic factors that define disease susceptibility. These studies are building on cell culture platforms that have been funded by the U.S. Department of Defense for their influenza studies. Finberg’s lab has developed a platform for studying the efficacy of antiviral approaches in cell culture and animal models, which will be applied, where helpful, to the novel SARS-CoV-2 therapies.

    Finberg’s lab works on the relationships between host cell surface proteins and viruses and bacteria, and the basis of cellular activation mediated by cell surface proteins. His clinical interests include immune-compromised hosts, infections in patients after transplant and inpatient infectious diseases.

  • Katherine Fitzgerald

    Katherine Fitzgerald, PhD

    Category: Host Response

    The Worcester Foundation for Biomedical Research Chair, professor of medicine and co-director of the Program in Innate Immunity

    Dr. Fitzgerald is researching, with Egil Lien, PhD, how the innate immune response to the novel coronavirus causes some individuals to die from an overwhelming cytokine storm, or the release of small proteins that affect cells’ interaction.

    The Fitzgerald lab is focused on understanding the molecular mechanisms controlling the inflammatory response. Her lab’s long-term goals are to understand how dysregulation of innate immunity underlies the development of infectious, inflammatory and autoimmune disease in humans.

    Fitzgerald Lab

    The Fitzgerald lab is part of the Program in Innate Immunity (PII) an interdisciplinary group of investigators who are focused on discovering the underlying mechanisms that drive immune defenses and inflammation, in both health and disease.

  • Guangping Gao

    Guangping Gao, PhD

    Category: Drug Design and Therapeutics

    The Penelope Booth Rockwell Professor in Biomedical Research, professor of microbiology & physiological systems, director of the Horae Gene Therapy Center and co-director of the Li Weibo Institute for Rare Diseases Research

    Dr. Gao is researching rAAV-based recombinant vaccines for COVID-19. The goal is to develop vectors that are capable of continually expressing soluble inhibitors of SARS-CoV-2. This would prevent the virus that causes COVID-19 from entering and infecting cells in the airway and lungs.

    An international leader in gene therapy research, Gao has played a key role in developing adeno-associated virus (AAV) gene therapies for a range of rare genetic disorders such as amyotrophic lateral sclerosis (ALS), Rett syndrome and Canavan disease.

    Gao Lab

  • Jonathan Gerber

    Jonathan Gerber, MD

    Category: Clinical Trials

    The Eleanor Eustis Farrington Chair in Cancer Research, associate professor of medicine and chief of the Division of Hematology/Oncology

    Dr. Gerber is leading a trial of convalescent plasma infusion to determine if it’s possible to give patients anti-SARS-CoV-2 antibody immunity passively, using plasma from a donor who has recovered from COVID-19.

    Gerber researches how to improve the identification and targeting of stem cells at the root of leukemia and related diseases, with the goal of personalizing therapy and developing treatments that are more effective and less toxic.

  • Anastasia Khvorova

    Anastasia Khvorova, PhD

    Category: Drug Design and Therapeutics

    Professor of RNA therapeutics

    The Khvorova lab is investigating a cocktail of siRNA and antisense RNA oligonucleotides that are specific for SARS-CoV-2, which in concept could be delivered to the macrophages and epithelial cells in the lungs and target the mRNA of the virus to significantly reduce replication of the virus in those cells.

    Dr. Khvorova is a leading expert in the biochemistry of RNA therapeutics. Scientists in the Khvorova lab identify the chemical and biological properties that drive small RNA tissue distribution, retention, cellular uptake and biological availability. Their goal is to deliver oligonucleotide and RNA-based therapies to tissues other than the liver through chemical engineering. By screening a wide range of chemically engineered and naturally occurring bioactive conjugates, they have already identified novel chemical modalities that support delivery of robust amounts of RNAs to the heart, kidneys, muscle, placenta, vasculature and brain—tissues previously not targeted by RNAi.

    Khvorova Lab

  • Mark Klempner

    Mark Klempner, MD

    Category: Drug Design and Therapeutics,Diagnostics and Vaccine Development

    Professor of medicine and executive vice chancellor of MassBiologics

    MassBiologics has previously demonstrated that human monoclonal antibodies are able to prevent infection with a related coronavirus, SARS CoV-1, and can be used for treatment of SARS-CoV-1 infections. Dr. Klempner has identified two human monoclonal antibodies that neutralize SARS-CoV-2 , including one that blocks binding of SARS-Cov-2 to its ACE2 receptor. Current efforts are directed at producing sufficient quantities of these antibodies for testing in an animal model of COVID-19.

    Another research effort at MassBiologics involves IgA antibodies, which are stabilized by the mucous that lines the respiratory tract. Klempner and colleagues are developing IgA antibodies for delivery directly to the respiratory tract for both prevention and treatment of COVID-19.

    Another approach being studied at MBL focuses on single-domain antibodies, also known as nanobodies—antibody fragments consisting of a single monomeric variable antibody domain that access unique sites on coronaviruses. Current efforts are directed at testing candidate nanobodies for cross neutralization and for in vivo testing against several coronavirus isolates.

    Diagnostics

    In collaboration with investigators at the New York Public Health laboratories at Wadsworth New York, MassBiologics has designed, engineered and manufactured three antigens associated with virus naturalizing antibodies.

  • Egil Lien

    Egil Lien, PhD

    Category: Host Response

    Professor of medicine

    Dr. Lien is researching, with Katherine Fitzgerald, PhD, how the innate immune response to the novel coronavirus causes some individuals to die from an overwhelming cytokine storm, or the release of small proteins that affect cell interaction. These approaches could lead to a better understanding of the critical illness response in profound lung injury and low blood pressure, which makes this virus fatal in some individuals.

    Lien’s research interests focus on how the body defends itself against disease-causing microbes by activation of the innate immune system. His lab also studies inflammation involved in the development of diabetes and obesity.

  • Shan Lu

    Shan Lu, MD, PhD

    Category: Diagnostics and Vaccine Development

    Professor of medicine

    Dr. Lu is investigating using a two-part, DNA primer vaccine with a protein boost for SARS-CoV-2.

    Lu is a pioneer in DNA vaccine development for HIV. The Lu lab has taken the novel approach of combining a DNA vaccine with a recombinant protein-based vaccine, such as the type used for human papillomavirus (HPV). In this approach, the HIV DNA acts as a primer to turn on the immune cells and is followed by a booster shot of the HIV protein vaccines, which stimulates production of the antibodies that fight the virus. This two-part approach activates both antibody and cell-mediated immune responses and induces functional antibodies with better qualities and longevity than using either type of vaccine alone.

  • Jeremy Luban

    Jeremy Luban, MD

    Category: Drug Design and Therapeutics

    The David J. Freelander Professor in AIDS Research and professor of molecular medicine

    The Luban lab is moving rapidly into the study and analysis of the virologic mechanism of COVID-19 infection, seeking to understand what makes its mechanism unique and why it might be susceptible to antiviral drugs like chloroquine. Dr. Luban’s 2016 paper on the singular infectivity of Ebola virus in Cell presaged the need for a more precise understanding of infectivity of novel viruses like SARS-CoV-2.

    Luban Lab

  • Katherine Luzuriaga

    Katherine Luzuriaga, MD

    Category: Clinical Trials

    The UMass Memorial Health Care Chair in Biomedical Research, professor of molecular medicine and director of the Center for Clinical and Translational Science

    Dr. Luzuriaga is leading an observational study of maternal-fetal transmission of SARS-CoV-2 and a trial of a pediatric antiviral drug.

    Luzuriaga researches how viruses, including human immunodeficiency virus (HIV) and Epstein Barr virus (EBV), establish persistent viral infections in children. She developed early diagnostic methods for pediatric HIV infection, led the first clinical trials of nevirapine in children, and conducted Phase I studies of several other antiretroviral therapies (ART) labeled for pediatric use. Having led the first early combination ART trials in infants, she is now focused on characterizing residual HIV reservoirs in individuals on ART to inform the development of strategies to achieve remission.

  • William McDougall

    William McDougall, PhD

    Category: Host Response

    Assistant professor of microbiology & physiological systems

    Dr. McDougall’s lab, funded by the federal Defense Advanced Research Projects Agency (DARPA), is applying tools designed to define critical host susceptibility factors for influenza infection to SARS-CoV-2 infection.

    McDougall joined the lab of Abraham Brass, MD, PhD, as a postdoctoral associate in 2014. His research includes the effect of viral infection on cell division, high-content screening of organelle biogenesis in humans using whole genome CRISPR libraries, and Chinese hamster ovary (CHO) whole genome CRISPR library construction and screen for increased production and antigenicity of HIV-SOSIP trimer.

    Brass Lab

  • Ann Moorman

    Ann Moorman, PhD, MPH

    Category: Host Response

    Professor of medicine

    Dr. Moormann is studying the age-specific differences in the immune responses and pathology of SARS-CoV-2 infection.

    Moormann's coronavirus work builds on her prior research on pediatric immunity to infectious diseases, including malaria and Epstein Barr virus, and how these infections lead to endemic Burkitt lymphoma, the most common pediatric cancer in sub-Saharan Africa. Her expertise covers immunology and infectious diseases, pediatrics, epidemiology and global health research.

  • Trudy Morrison

    Trudy Morrison, PhD

    Category: Diagnostics and Vaccine Development

    Professor of microbiology & physiological systems

    Dr. Morrison is developing and testing vaccine candidates for COVID-19 by adapting virus-like particles (VLP) she previously found to treat respiratory syncytial virus, or RSV. She is also developing multivalent self-assembling nanoparticles that can trigger the formation of antibodies in patients to prevent the coronavirus from infecting cells. These nanoparticles may conceivably be administered either through the nose or via an inhaler like those used to deliver asthma medication.

    The Morrison lab explores how paramyxovirus—a group of RNA viruses that cause acute respiratory diseases that include mumps, measles and RSV—gain entry into cells and assemble and release virus. Morrison investigates the protein interactions required for the formation of assembly complexes, the cell domains involved in virus assembly, and the host cell contributions to virus assembly and release.

  • James Munro

    James Munro, PhD

    Category: Diagnostics and Vaccine Development

    Associate professor of microbiology & physiological systems

    Dr. Munro is conducting basic virology studies on the interactions between SARS-CoV-2 and cells in the airway and lungs. He is exploring the processing and presentation of viral antigens, which could reveal biological keys for designing protective vaccines.

    The Munro lab is devoted to the development and application of biophysical approaches to studying numerous viral pathogens, including HIV, influenza, Ebola and Lassa. Munro’s work incorporates elements of virology, biophysics, biochemistry and computational modeling.

    Munro Lab

  • Celia Schiffer

    Celia Schiffer, PhD

    Category: Drug Design and Therapeutics

    The Gladys Smith Martin Chair in Oncology, professor of biochemistry & molecular pharmacology and director of the Institute for Drug Resistance

    Dr. Schiffer is collaborating with Paul Thompson, PhD, to identify and design antiviral drugs for SARS-CoV-2, using high-throughput drug screening and innovative structure-based drug design.

    Schiffer studies how viruses become resistant to drugs and uses a variety of research tools to determine how changes in the biochemistry of a drug can make resistance less likely.

    Schiffer Lab

  • Miguel Sena-Esteves

    Miguel Sena-Esteves, PhD

    Category: Diagnostics and Vaccine Development

    Associate professor of neurology

    Collaborating with Trudy Morrison, PhD, the Sena-Esteves lab is testing the potential of virus-like particles (VLP) as vaccine candidates for COVID-19.

    Dr. Sena-Esteves designs and engineers AAV vectors that carry therapeutic genes to the central nervous system to treat fatal neurological diseases. The Sena-Esteves lab is investigating new AAV treatments for lysosomal storage diseases such as GM1-gangliosidosis and GM2-gangliosidoses (Tay-Sachs and Sandhoff diseases).

    Sena-Esteves Lab

  • Paul Thompson

    Paul Thompson, PhD

    Category: Drug Design and Therapeutics

    Professor of biochemistry & molecular pharmacology and director of the Chemical Biology Interface Program

    Dr. Thompson is collaborating with Celia Schiffer, PhD, to identify and design antiviral drugs for SARS-CoV-2, using high-throughput drug screening and innovative structure-based drug design. One area of particular promise is the identification and development of protease inhibitors specific to the virus. Thompson’s lab has vast experience in small-molecule screening, where researchers test thousands of potential therapeutics against infectious agents like viruses. Thompson’s identification of a family of enzymes critical to the development of autoimmune diseases helped advance the understanding of the biochemistry and biological effects of these enzymes.

    Thompson Lab

  • Jonathan Watts

    Jonathan Watts, PhD

    Category: Drug Design and Therapeutics

    Associate professor of RNA therapeutics

    Scientists in the Watts lab are developing viable mechanisms for delivering small interfering RNAs (siRNA) to the lungs, which can significantly reduce the expression of the SARS-CoV-2 viral genes.

    The Watts lab is developing new approaches to several classes of oligonucleotides with the goal of harnessing the chemistry and biology of these molecules to help make oligonucleotides an effective therapeutic for disease. Oligonucleotides are short DNA or RNA molecules that have a wide range of applications. By synthesizing oligonucleotides that can bind to the messenger RNA, it may be possible to effectively turn off genes that cause illness and disease.

    Watts Lab

  • Scot Wolfe

    Scot Wolfe, PhD

    Category: Host Response

    Professor of molecular, cell & cancer biology

    Dr. Wolfe is working with Robert W. Finberg, MD, on studies using CRISPR/Cas9 gene editing tools to define the virus and host genetic factors that define disease susceptibility.

    Wolfe’s research interests focus on engineering precise gene editing systems for application in gene therapy and the analysis of gene regulatory networks.

    Wolfe Lab

  • Jun Xie

    Jun Xie, PhD

    Category: Drug Design and Therapeutics

    Associate professor of microbiology & physiological systems

    Dr. Xie, collaborating with Guangping Gao, PhD, is developing rAAV-based recombinant vaccines for COVID-19. Xie researches AAV vector technology and AAV-mediated miRNA therapeutics.