Skip to main content

Author: The Wistar Institute

Wistar Appoints Ami Patel, Ph.D., as Caspar Wistar Fellow

PHILADELPHIA — (Sept. 14, 2020) — The Wistar Institute, an international biomedical research leader in cancer, immunology, infectious disease research, and vaccine creation, announces the appointment of Ami Patel, Ph.D., as a Caspar Wistar Fellow in the Vaccine and Immunotherapy Center.

Dr. Patel’s research focuses on strategies to combat emerging infectious diseases, including engineering vaccines and immunotherapies against viral and bacterial pathogens. Her independent program will expand Wistar’s expertise in pandemic preparedness and response to emerging outbreaks.

As a Caspar Wistar Fellow, Dr. Patel will embark on an independent path where her lab will harness the human immune system using next-generation technologies to improve public health.

“Emerging pathogens are of deep interest to me, as my work on the avian flu, swine flu, Ebola, and the current COVID-19 pandemic have demonstrated,” said Patel. “As an infectious disease scientist, I always ask myself ‘What will be the next outbreak, what do we need to understand to quicken our response and how can we help our bodies fight the pathogen?’ The COVID-19 pandemic is the perfect example of why reaction time to halt an outbreak matters and how effective technologies and resources are needed to develop vaccines. The Caspar Wistar Fellowship gives me the opportunity to pursue my own research ideas while still enjoying a lot of hands-on research and benefiting from mentoring in the highly collaborative environment at Wistar.”

The Caspar Wistar Fellows Program nurtures outstanding young scientists during their critical early years as independent investigators, creating a springboard for them to launch their careers and join the next generation of scientific leaders. The Program was made possible by the support of Wistar Board of Trustees member Doug Briggs and his wife Peggy.

“As a Wistar supporting partner, it is inspiring to see a promising early-career scientist that is so clear in her research goals and steadfast in her pursuit of biomedical innovation excellence,” said Doug Briggs. “Peggy and I could not be more pleased to see our second Caspar Wistar Fellow fast-tracked to reach her potential and beyond. We believe Dr. Patel will go on to do great things in science.”

Prior to her latest appointment, Patel conducted postdoctoral research at the San Raffaele Telethon Institute for Gene Therapy, Milan, Italy, and at Wistar in the lab of David Weiner, Ph.D. She received a Ph.D. in medical microbiology from the University of Manitoba, Winnipeg, Canada. Her doctoral research was performed in the Special Pathogens Program at the Public Health Agency of Canada, Winnipeg, Canada.

“Both mentorship and freedom to pursue a strong, independent research program are the key components to this Program,” said Dario Altieri, M.D., Wistar president and CEO. “I’m delighted that our quest to find the best and brightest led us to Dr. Ami Patel. Her research strengths in novel engineering approaches for in vivo gene delivery using DNA platforms for emerging infectious diseases and antimicrobial-resistant bacteria are a perfect asset as we build-out our pandemic response team.”

###

The Wistar Institute is an international leader in biomedical research with special expertise in cancer, immunology, infectious diseases and vaccine development. Founded in 1892 as the first independent nonprofit biomedical research institute in the United States, Wistar has held the prestigious Cancer Center designation from the National Cancer Institute since 1972. The Institute works actively to ensure that research advances move from the laboratory to the clinic as quickly as possible. wistar.org.

Scientists Engineer DNA-based Nanotechnology to Stimulate Potent Antitumor Immune Responses in Preclinical Models

PHILADELPHIA — (Sept. 10, 2020) — Combining their expertise in protein engineering and synthetic DNA technology, scientists at The Wistar Institute successfully delivered nanoparticle antitumor vaccines that stimulated robust CD8 T cell immunity and controlled melanoma growth in preclinical models. Study results were published online in Cancer Immunology Research, a journal of the American Association for Cancer Research, and support exploration of this immunotherapy approach for additional cancer types.

Nanovaccines consist of extremely small (nano) particles — similar in size to bacteria and viruses — used to display multiple copies of an antigen and able to elicit strong immune responses. The team previously reported on using DNA instructions to launch in vivo production of nanoparticle vaccines (DLnano-vaccines).

DLnano-vaccines assembled in the body produced stronger immune responses than protein based nanoparticle vaccines in an infectious disease setting, especially inducing CD8 T cell responses.

“We wanted to test DLnano-vaccines for cancer immunotherapy and obtain proof of concept that this platform could be successfully applied in the cancer field, thanks to its effectiveness at prompting CD8 T cells responses,” said Daniel Kulp, Ph.D., associate professor in Wistar’s Vaccine & Immunotherapy Center and co-corresponding author of the study, who specializes in nanotechnology and protein engineering for vaccine development.

Due to their ability to specifically kill malignant cells, CD8 T cells play a pivotal role in anticancer immunity, therefore engagement of these cells represents a necessary step for the success of anticancer vaccine approaches, although this type of immune response is typically difficult to achieve by vaccination with proteins or inactivated virus.

Researchers designed DLnano-vaccines displaying 60 copies of protein parts derived from the melanoma-specific antigens Trp2 and Gp100 and tested these in mouse models of melanoma, observing prolonged survival that depended on CD8 T cell activation both in therapeutic and prophylactic settings.

“One of the advantages of synthetic DNA technologies over other methods is the versatility of the platforms,” said Ziyang Xu, Ph.D., a recent doctoral graduate working at Wistar and the first author of the study. “DLnano-vaccines may be designed for various cancer targets and our study shows this is a promising strategy for cancer immunotherapy that may warrant further testing.”

To elucidate the mechanism through which DLnano-vaccines activate CD8 T cells, the team studied the effects of the DNA-launched version of a previously described HIV nanoparticle vaccine (eOD-GT8-60mer). They observed that DLnano-vaccines administered via electroporation resulted in transient muscle cell apoptosis that attracted macrophage infiltration at the injection site, which in turn was instrumental to activate CD8 T cells.

DLnano-vaccines were developed using synthetic DNA technology in collaboration with the lab of David B. Weiner, Ph.D., Wistar executive vice president, director of the Vaccine & Immunotherapy Center, and the W.W. Smith Charitable Trust Professor in Cancer Research and also a co-senior author on the study.

Co-authors: Neethu Chokkalingam, Edgar Tello-Ruiz, Mamadou A. Bah, Susanne Walker, and Nicholas J. Tursi from Wistar; Megan C. Wise, Paul D. Fisher, Katherine Schultheis, Kate E. Broderick, and Laurent Humeau from Inovio Pharmaceuticals, Inc.

Work supported by: National Institutes of Health (NIH) grants U19 Al109646 and Collaborative Influenza Vaccine Innovation Centers (CIVICs) contract 75N93019C00051; additional support was provided by Inovio Pharmaceuticals, Inc., a grant from the W.W. Smith Charitable Trust, and the Monica H.M. Shander Memorial Fellowship.

Publication information: A DNA-launched nanoparticle vaccine elicits CD8+ T-cell immunity to promote in vivo tumor control, Cancer Immunology Research, 2020. Online publication.

###

The Wistar Institute is an international leader in biomedical research with special expertise in cancer, immunology, infectious disease research, and vaccine development. Founded in 1892 as the first independent nonprofit biomedical research institute in the United States, Wistar has held the prestigious Cancer Center designation from the National Cancer Institute since 1972. The Institute works actively to ensure that research advances move from the laboratory to the clinic as quickly as possible. Wistar’s Business Development team is dedicated to accelerating the translation of Wistar discoveries into innovative medicines and healthcare solutions through licensing, start-ups and creative collaborations. wistar.org.

The BEAT-HIV Martin Delaney Collaboratory Issues Recommendations on Measuring Persistent HIV Reservoirs in Cure-directed Clinical Trials

PHILADELPHIA — (Sept. 7, 2020) — Spearheaded by scientists at The Wistar Institute, top worldwide HIV researchers from the BEAT-HIV Martin Delaney Collaboratory to Cure HIV-1 Infection by Combination Immunotherapy (BEAT-HIV Collaboratory) compiled the first comprehensive set of recommendations on how to best measure the size of persistent HIV reservoirs during cure-directed clinical studies. This perspective article was published today in Nature Medicine.

Cure-directed studies seek to control or eradicate HIV beyond current antiretroviral therapy (ART) which can only suppress but not eliminate HIV. Long-term viral persistence on ART continues to cause immune activation, chronic inflammation and progressive damage to multiple organs. Multiple cure-directed studies are underway worldwide but no consensus statement was available to prioritize and interpret the many strategies available today to measure persistent HIV on ART.

“Bringing together many of the original investigators who developed current assays used to measure HIV, the BEAT-HIV Collaboratory has now issued recommendations for priority in HIV measures as a guide for cure-directed studies,” said Luis J. Montaner, D.V.M., D.Phil., the Herbert Kean, M.D., Family Professor and director of the HIV-1 Immunopathogenesis Laboratory at Wistar’s Vaccine & Immunotherapy Center, co-leader of the Delaney Collaboratory and corresponding author on the article. “A major obstacle to eradication is the virus hiding in some compartments of the immune system where it’s difficult to target and measure. The BEAT-HIV guidelines now provide specific information on the strengths and limitations of each assay available today.”

The ability to accurately measure the size of these HIV reservoirs is critical when evaluating potential therapeutic strategies to cure HIV. It is also necessary for monitoring viral levels and guide ART interruption.

“We systematically reviewed the state of the science in the field and provided a collective and comprehensive view on which viral measurements to prioritize in clinical trials,” said Mohamed Abdel-Mohsen, Ph.D., assistant professor in Wistar’s Vaccine & Immunotherapy Center and one of the authors of the paper. “I think this is a crucial step to take the best advantage of the most valuable resource available to researchers in their quest to find a cure for HIV, the blood and tissue samples from people living with HIV who generously participate in the HIV cure-focused clinical trials all over the world.”

In current HIV cure-directed studies in ART-suppressed people living with HIV, viral levels are monitored in peripheral blood cells obtained either by phlebotomy or leukapheresis (a laboratory procedure to separate white blood cells from whole blood) and biopsies from gut-associated lymphoid tissue or lymph nodes, though most trials only use peripheral blood because it is easier to collect.

In this work, the BEAT-HIV consortium compiled priority lists of the available assays and technologies to apply for each specimen type and collection method with the aim of aiding standardization of cure-directed trials.

Co-authors: Douglas Richman and David M Smith from VA San Diego Healthcare System and University of California, San Diego; Robert F. Siliciano and Janet D. Siliciano from Johns Hopkins University; Michel C. Nussenzweig, Christian Gaebler and Marina Caskey from The Rockefeller University; Bonnie Howell and Daria Hazuda from Merck & Co; Javier Martinez-Picado from IrsiCaixa AIDS Research Institute, Badalona, Spain, University of Vic – Central University of Catalonia, Spain and Catalan Institution for Research and Advanced Studies (ICREA), Spain; Nicolas Chomont from Université de Montréal, Canada; Katharine J Bar, Frederic Bushman, Michael R. Betts, Beatrice H. Hahn, Ian Frank, James L Riley, and Pablo Tebas from University of Pennsylvania; Xu G. Yu and Mathias Lichterfeld from Ragon Institute of MGH, MIT and Harvard and Brigham and Women’s Hospital; Jose Alcami and Maria J. Buzon from Instituto de Salud Carlos III, Madrid and Infectious Diseases Unit, IBIDAPS, Hospital Clinic, University of Barcelona, Spain; Adam M. Spivak and Vicente Planelles from University of Utah; Ya-Chi Ho from Yale School of Medicine; Mirko Paiardini from Yerkes National Primate Research Center and Emory University; Qingsheng Li from School of Biological Sciences and Nebraska Center for Virology, University of Nebraska-Lincoln; Jacob D. Estes from Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health and Science University; Thomas J Hope from Northwestern University; Jay Kostman and Karam Mounzer from Jonathan Lax Center, Philadelphia FIGHT; and Lawrence Fox from Division of AIDS, NIAID, NIH. All authors contributed to the writing and editing of the manuscript.

Wistar authors were supported by: National Health Institutes (NIH)-funded BEAT-HIV Martin Delaney Collaboratory to cure HIV-1 infection 1UM1Al126620; NIH grants R01 AI065279, U01 AI065279, R01 DA048728, R01 DA049666, R01 DK123733, R01 AG062383, R01NS117458, R21 AI143385, R21 AI129636, and R21 NS106970; Herbert Kean, M.D., Family Endowed Chair Professorship; the Robert I. Jacobs Fund of the Philadelphia Foundation; amfAR, The Foundation for AIDS Research impact grant 109840-65-RGRL; W.W. Smith Charitable Trust grant A1901; Wistar Cancer Center Support Grant P30 CA010815-49S2; and the Penn Center for AIDS Research P30 AI 045008.

Publication information: Recommendations for Measuring HIV Reservoir Size in Cure-directed Clinical Trials, Nature Medicine, 2020. Online publication.

###

The Wistar Institute is an international leader in biomedical research with special expertise in cancer, immunology, infectious disease research, and vaccine development. Founded in 1892 as the first independent nonprofit biomedical research institute in the United States, Wistar has held the prestigious Cancer Center designation from the National Cancer Institute since 1972. The Institute works actively to ensure that research advances move from the laboratory to the clinic as quickly as possible. Wistar’s Business Development team is dedicated to accelerating the translation of Wistar discoveries into innovative medicines and healthcare solutions through licensing, start-ups and creative collaborations. wistar.org.

Scientists Use Knowledge From Traditional Medicine to Discover Drugs to Fight the Pandemic

Dr. Ian Tietjen came to Wistar in 2019 as a research assistant professor in the laboratory of Dr. Luis Montaner. He brings a fascinating approach to HIV and infectious disease research that explores the traditional medicine knowledge of Indigenous peoples and healers around the world in treating infection with natural substances.

“There is a wealth of information and experience that has been passed down through different cultures over generations and allowed people to maintain health and treat disease before the advent of modern medicine,” said Tietjen. “While a lot of this information can be anecdotal and sometimes inexplicable to scientists like us, we believe it could hold the key to powerful medicines. We want to apply our scientific method and currently available research technologies to find out.”

Many natural compounds have provided antiviral activity during evolution, for example to protect plants and other organisms that don’t have a specialized immune system like ours and use chemicals to fight viruses. Therefore, these substances could be repurposed as antivirals to support our immune response against infection.

Based on this idea, botanists and chemists began teaming up in the 1940s to create antiviral drugs using chemical compounds found in plants, and during the past 25 years, several screening programs have investigated the antiviral activity of medicinal plants.

Dr. Tietjen brought to Wistar a collection of compounds derived from plant and marine organisms that he obtained in collaboration with other researchers around the world and while working with traditional healers in different countries. The Montaner lab is now testing these substances for their ability to affect the immune response to HIV.

When COVID-19 started, Drs. Tietjen and Montaner decided to apply the same approach and comb through the library to look for antiviral activity against SARS-CoV-2. For example, they are testing for the ability of these compounds to disrupt the interaction between the viral Spike protein and the ACE2 receptor present on human cells — the lock and key mechanism that allows virus entry — in order to halt infection.

“So far, we’ve found several pure compounds that are able to do so in the laboratory,” said Tietjen. “We also identified several African medicinal plant extracts that disrupt this interaction.”

This was accomplished in collaboration with Joel Cassel in Wistar’s Molecular Screening Facility. Cassel successfully adapted a pre-existing assay for large-scale screening, which allowed the team to evaluate many compounds and find positive hits quickly. To confirm the screening results, they tested those hits against a different receptor/viral protein interaction, and many of them didn’t show activity in this experiment, indicating they specifically block the SARS-CoV-2 entry mechanism.

“Importantly, we are observing similar bioactivities in specimens of the same plant collected from different healers and in different locations, which supports that the bioactivity is intrinsic to the plant,” said Tietjen.

Some of the medicinal plant extracts are extremely potent, in some cases disrupting the Spike/ACE2 interaction at very low concentrations.

“One of our leads includes a species of Artemisia, a family of plants commonly known as mugwort, wormwood and sagebrush and well established as an antimalarial agent. Our observation is consistent with what others are reporting worldwide, although it’s way too early to recommend use of this plant to treat COVID-19,” said Tietjen.

He added that some healers in Southern Africa have administered some of the medicinal plants being tested at Wistar to community members that have been coming to them recently with respiratory infections, and they report having obtained benefits.

“This information is anecdotal, so we have to discuss our findings together and all agree on a plan to monitor these observations more closely,” Tietjen said. “This is the perfect example of the benefits of what we call ‘reverse pharmacology’ efforts — we obtain compounds and information by collaborating with traditional healers, test their products in the lab with our methods and share our results with them as the data are generated to inform their practices in real-time.”

Tietjen also emphasizes that collaborations with healers and their communities involves years of building relationships and trust. It also requires working together as equal partners and toward mutual knowledge benefits. “This exchange, being able to provide this kind of information to healers, and also learning about quite different but also valid ways to perform medicine are very rewarding aspects of this work,” he continued. “We can learn a lot from each other.”

In the lab, researchers are now confirming preliminary results using a SARS-CoV-2 pseudovirus — virus-like particles that have the essential components for infecting cells in culture but can’t replicate and cause further infection, representing safe tools to mimic SARS-CoV-2 virus for further studies.

Tietjen, Montaner and team are also working to determine if the hit compounds can block entry of the real SARS-CoV-2 into cells in vitro. This work can be done at Wistar in the BSL3 facility, which provides the required safety environment for handling airborne infectious diseases like coronaviruses.

Next, researchers plan to refine and optimize the most potent molecules to make them as selective and safe as possible.

A longer-term goal is to identify agents that can enhance the antiviral activity of existing drugs like remdesivir, along with those that could inhibit entry of other coronaviruses that use ACE2 for entry. Those leads would eventually go into animal models to test for in vivo efficacy.

“Our work looking for natural antivirals is worthwhile because, even when a SARS-CoV-2 vaccine becomes available, it won’t be accessible to everyone immediately, especially in resource-limited regions where people rely on traditional medicines and community healers for their health,” said Tietjen. “Also, since the world has been hit by different coronavirus outbreaks since 2003, it’s critical for us to build up our antiviral armamentarium and be prepared for the next potential coronavirus threat to human health.”

Wistar Science Highlights: New Discoveries on HIV Latency and How Cancer Cells Talk With Their Neighboring Normal Cells

Antiretroviral therapy (ART) has dramatically increased the health and life expectancy of HIV-infected individuals, suppressing virus replication in the host immune cells and stopping disease progression; however, low yet persistent amounts of virus remain in the blood and tissues despite therapy. Virus persistency limits immune recovery and is associated with chronic levels of inflammation so that treated HIV-infected individuals have higher risk of developing a number of diseases.

This persistent infection stems from the ability of HIV to hide in a rare population of CD4 T cells. Finding new markers to identify the virus reservoir is of paramount importance to achieve HIV eradication.

The lab of Mohamed Abdel-Mohsen, Ph.D., assistant professor in the Vaccine & Immunotherapy Center, may have discovered a new way of identifying and targeting hidden HIV reservoirs during ART.

The sugar molecules present on the surface of immune cells play a critical role in regulating their functions and fate. Researchers explored the role of the sugar component on the surface of host cells and described a “glycomic — or sugar — signature” that can impact HIV persistence.

Published in Cell Reports, the findings may have translational implications for improving the long-term care of HIV positive people.


The crosstalk between cancer cells and their neighboring normal cells is important to promote cancer progression.

The lab of Dario C. Altieri, M.D., Wistar president and CEO, director of the Institute’s Cancer Center and the Robert & Penny Fox Distinguished Professor, studied how this exchange happens to gain more clues on how tumors “hijack” their microenvironment and promote disease progression and recurrence.

Researchers cultured breast cancer cells in low-oxygen to mimic a condition known as hypoxia, which is a hallmark of the microenvironment surrounding most solid tumors. In this setting, they discovered that cancer cells package oncogenic messages into tiny particles called extracellular vesicles and send them to neighboring normal epithelial cells. This results in reprogramming of the shape and position of their mitochondria — the cell’s powerhouse — to ultimately alter tissue structure.

These findings, published in the journal Developmental Cell, suggest novel therapeutic targets to disrupt the pro-tumorigenic changes caused by cancer cells to the microenvironment.

Breast Cancer Cells Use Message-carrying Vesicles to Send Oncogenic Stimuli to Neighboring Normal Cells 

PHILADELPHIA — (Aug. 10, 2020) — According to a study by The Wistar Institute, breast cancer cells starved for oxygen send out messages that induce oncogenic changes in surrounding normal epithelial cells. These messages are packaged into particles called extracellular vesicles (EVs) and reprogram mitochondrial shape and position within the recipient normal cells to ultimately promote deregulated tissue morphogenesis. These findings were published today in Developmental Cell.

“It is well known that cancer cells ‘talk’ to their neighboring normal cells all the time and this is important to promote cancer progression,” said study lead author Dario C. Altieri, M.D., Wistar president and CEO, director of the Institute’s Cancer Center and the Robert & Penny Fox Distinguished Professor. “How that happens and what signals are being transferred from one cell to another are still very much open questions. A better understanding of this process may give us important clues about how tumors hijack nearby normal cells to promote disease recurrence.”

For their studies, Altieri’s team cultured breast cancer cells in a low-oxygen setting to mimic a condition known as hypoxia, which is a hallmark of the microenvironment surrounding most solid tumors, and studied the EVs released by these cells.

EVs are tiny structures enclosed in a double membrane layer and released by most cells to transfer different molecules and information to other cells. As such, vesicles are an important means of intercellular communication. In this study, researchers focused on small EVs (sEV) that are between 30 and 150 nm in size.

To dissect the effects of sEVs produced by cancer cells on normal neighboring cells, researchers incubated normal breast epithelial cells with sEVs released by cells maintained in hypoxia. They observed an increase in the ability of normal recipient cells to migrate in culture, which in turn correlated with a redistribution of their mitochondria to the cell periphery. This is consistent with the role played by mitochondria in supporting cell motility, previously described by the Altieri lab.

In addition to modulation of mitochondrial behavior, the research team discovered that sEV released by hypoxic breast cancer cells induced major changes in gene expression in the normal recipient cells, with activation of multiple pathways of cell motility, cytoskeletal organization and cell-to-cell contact. Additionally, sEV-treated cells exhibited reduced cell death and increased pro-inflammatory responses.

Altieri and colleagues went on to identify Integrin-Linked Kinase (ILK) as the main signaling component packaged in sEVs, responsible for both mitochondrial changes and increased migration of recipient cells.

In turn, activation of ILK signaling profoundly affected normal tissue morphogenesis. Using 3-D cell models of normal mammary gland development, the team observed that exposure to sEVs from hypoxic cancer cells caused a general disruption of the normal mammary gland architecture and induced multiple traits of oncogenic transformation, including morphological changes, deregulated cell proliferation, reduced cell death, and appearance of markers of epithelial-mesenchymal transition (EMT), a process that confers mobility to cancer cells and the capacity to migrate from the primary site.

“Our findings indicate that breast cancer cells may use sEVs to enable both local and distant disease progression,” said Irene Bertolini, Ph.D., first author of the study and a postdoctoral fellow in the Altieri lab. “Based on these observations, we suggest that therapeutic targeting of ILK or mitochondrial reprogramming may provide novel strategies to disrupt these pro-tumorigenic changes in the microenvironment.”

Co-authors: Jagadish C. Ghosh and Andrew V. Kossenkov from Wistar; Sudheer Mulugu from Perelman School of Medicine, University of Pennsylvania; Shiv Ram Krishn and Lucia R. Languino from Sidney Kimmel Cancer Center, Thomas Jefferson University; Valentina Vaira from Fondazione IRCCS Caʹ Granda Ospedale Maggiore Policlinico, Milan, Italy and University of Milan, Italy; and Jun Qin and Edward F. Plow from Lerner Research Institute, Cleveland Clinic.

Work supported by: National Institutes of Health (NIH) grants P01 CA140043, R35 CA220446, R01 HL58758, and R50 CA211199; Additional support was provided by the Italian Minister of Health. Core support for The Wistar Institute was provided by the Cancer Center Support Grant P30CA010815.

Publication information: Small Extracellular Vesicle Regulation of Mitochondrial Dynamics Reprograms a Hypoxic Tumor Microenvironment, Developmental Cell (2020). Online publication.

###

The Wistar Institute is an international leader in biomedical research with special expertise in cancer, immunology, infectious disease research, and vaccine development. Founded in 1892 as the first independent nonprofit biomedical research institute in the United States, Wistar has held the prestigious Cancer Center designation from the National Cancer Institute since 1972. The Institute works actively to ensure that research advances move from the laboratory to the clinic as quickly as possible. Wistar’s Business Development team is dedicated to accelerating the translation of Wistar discoveries into innovative medicines and healthcare solutions through licensing, start-ups and creative collaborations. wistar.org.

New Sugar-based Signature Identifies T Cells Where HIV Hides and Persists Despite Antiretroviral Therapy

PHILADELPHIA — (August 4, 2020) — Scientists at The Wistar Institute may have discovered a new way of identifying and targeting hidden HIV viral reservoirs during treatment with antiretroviral therapy (ART). These findings were published today in Cell Reports and may have translational implications for improving the long-term care of HIV positive people.

ART has dramatically increased the health and life expectancy of HIV-infected individuals, suppressing virus replication in the host immune cells and stopping disease progression; however, low yet persistent amounts of virus remain in the blood and tissues despite therapy. Virus persistency limits immune recovery and is associated with chronic levels of inflammation so that treated HIV-infected individuals have higher risk of developing a number of diseases.

This persistent infection stems from the ability of HIV to hide in a rare population of CD4 T cells. Finding new markers to identify the virus reservoir is of paramount importance to achieve HIV eradication.

“With recent advances that we are making in the fields of glycobiology and glycoimmunology, it has become clear that the sugar molecules present on the surface of immune cells play a critical role in regulating their functions and fate,” said corresponding author Mohamed Abdel-Mohsen, Ph.D., assistant professor in The Wistar Institute Vaccine & Immunotherapy Center. “However, the relevance of host cell-surface glycosylation in HIV persistence remained largely unexplored, making it a ‘dark matter’ in our understanding of HIV latency. For the first time, we described a cell-surface glycomic signature that can impact HIV persistence.” Persistently infected cells can be divided into two groups: cells where the virus is completely silent and does not produce any RNA (i.e., silent HIV reservoir); and cells where the virus produces low levels of RNA (i.e., active HIV reservoir). Targeting and eliminating both types of reservoirs is the focus of the quest for an HIV cure. A main challenge in this quest is that we do not have a clear understanding of how these two types of infected cells are different from each other and from HIV-uninfected cells. Therefore, identifying markers that can distinguish these cells from each other is critical.

For their studies, Abdel-Mohsen and colleagues used a primary cell model of HIV latency to characterize the cell-surface glycomes of HIV-infected cells. They confirmed their results in CD4 cells directly isolated from HIV-infected individuals on ART.

They identified a process called fucosylation as a feature of persistently infected T cells in which the viral genome is actively being transcribed. Fucosylation is the attachment of a sugar molecule called fucose to proteins present on the cell surface and is critical for T-cell activation.

Researchers also found that the expression of a specific fucosylated antigen called Sialyl-LewisX (SLeX) identifies persistent HIV transcription in vivo and that primary CD4 T cells with high levels of SLeX have higher levels of T-cell pathways and proteins known to drive HIV transcription during ART. Such glycosylation patterns were not found on HIV-infected cells in which the virus is transcriptionally inactive, providing a distinguishing feature between these two cell compartments. Interestingly, researchers also found that HIV itself promotes these cell-surface glycomic changes.

Importantly, having a high level of SLeX is a feature of some cancer cells that allow them to metastasize (spread to other sites in the body). Indeed, researchers found that HIV-infected cells with high levels of SLeX are enriched with molecular pathways involved in trafficking between blood and tissues. These differential levels of trafficking might play an important role in the persistence of HIV in tissues, which are the main sites where HIV hides during ART.

Based on these findings, the role of fucosylation in HIV persistence warrants further studies to identify how it contributes to HIV persistence and how it could be used to target HIV reservoirs in blood and tissues.

Co-authors: Co-first authors are Florent Colomb, Leila B. Giron, and Opeyemi S. Adeniji from Wistar and Leticia Kuri-Cervantes from University of Pennsylvania; Other authors include Harsh Dweep and Andrew V. Kossenkov from Wistar; Michael R. Betts from University of Pennsylvania; Tongcui Ma and Nadia R. Roan from University of California San Francisco and Gladstone Institutes; Emilie Battivelli and Eric Verdin from The Buck Institute for Research on Aging; Clovis S. Palmer from The Burnet Institute, Australia; and Hiroaki Tateno from National Institute of Advanced Industrial Science and Technology, Japan.

Work supported by: National Health Institutes (NIH) grants R01 DK123733, R01 AG062383, R01 NS117458, R21 AI143385, R21 AI129636, and R21 NS106970; the Penn Center for AIDS Research; The Foundation for AIDS Research (amfAR) impact grant 109840-65-RGRL. Additional support was provided by the NIH-funded BEAT-HIV ‘Martin Delaney Collaboratory to cure HIV-1 infection’, the Kean Family Professorship, and the Robert I. Jacobs Fund of the Philadelphia Foundation.

Publication information: Sialyl-Lewis-XGlyco-antigen Is Enriched on Cells with Persistent HIV Transcription During Therapy, Cell Reports, 2020. Online publication.

###

The Wistar Institute is an international leader in biomedical research with special expertise in cancer research and vaccine development. Founded in 1892 as the first independent nonprofit biomedical research institute in the United States, Wistar has held the prestigious Cancer Center designation from the National Cancer Institute since 1972. The Institute works actively to ensure that research advances move from the laboratory to the clinic as quickly as possible. wistar.org.

Workforce Development at Wistar: Life Science Students Gain Valuable Research Experience During the COVID-19 Pandemic

As our state economy faces a long slog back to recovery from the COVID-19 pandemic, Wistar is poised to assist with the development of the workforce that will help enable that recovery. The Institute’s education and training programs provide just what is needed for curious, intellectually minded students to establish careers as biomedical research technicians and pursue other careers in the life sciences. 

The Biomedical Technician Training (BTT) Program and the Biomedical Research Technician (BRT) Apprenticeship focus on giving students an accelerated career path with on-the-job training as biomedical research technicians.

The BTT Program was launched 20 years ago to develop the workforce in the biomedical and biopharmaceutical research sectors and provide hands-on training to students for two years at the lab bench to become research technicians. The experience brings science vividly to life and inspires many students to pursue advanced degrees or develop highly specialized biotechnology research skills through Wistar’s state-certified, non-traditional BRT Apprenticeship.

The BRT Apprenticeship gives interested BTT Program graduates nine additional months of intensive experience, building critical research skills and specialization through training in top biomedical research labs across the region. In 2019, the BRT Apprenticeship was awarded the Outstanding Non-traditional Apprenticeship Program of the year by the Pennsylvania Commonwealth’s Department of Labor & Industry. 

Wistar’s education and research training programs boast many stories of talented trainees who have matured into skilled scientists. Some of the BTT and BRT trainees are currently involved in laboratories working to understand more about COVID-19, having the opportunity to make a difference during this global health crisis.

Yaya Dia is one of them. He works in the Wistar lab of Dr. David Weiner and contributes to translational research taking place to advance a COVID-19 synthetic DNA vaccine which is being tested in clinical trials. Yaya trained in the Weiner lab during his BTT internship and is currently a BRT apprentice working in the lab on various projects and contributing to the vital research that led to his name added as a co-author on four papers. 

“It is a pleasure and privilege to work on the DNA vaccine that Dr. Weiner and my colleagues are advancing,” said Yaya. “I was in the right place at the right time to have this amazing experience. I carry out assays and tests to better understand the immune system and how it reacts to certain pathogens. I love that I get to better understand how the body works and how it fights diseases with the help of a DNA vaccine. It’s a great lab and a great team and Dr. Weiner really cares about the people he works with, which makes you want to work harder.” Yaya sees his next career step in science enrolling into a Physician Assistant program or medical school.

Jessicamarie Morris and Kwasi Gyampoh are BRT Apprenticeship graduates who work in the Wistar lab of Dr. Luis Montaner, who has been focusing on developing a cure for HIV and now is expanding his research activities to include discovering new therapies for COVID-19. With Dr. Montaner as their mentor, they have become sought-after and valued biomedical research technicians.

“I would have never been where I am without either program,” said Jessicamarie. “I had graduated college and felt I was done when it came to more schooling. I thought that traditional route was the only way to get into the science field. But I realized the BTT Program and the BRT Apprenticeship offer a novel entrance into science.”

She learned by doing and that paid off in spades.

“At Wistar, I experienced what it means to work in a real, functioning lab within a research institute. I was able to do so many different types of work and learned the ins and outs of different departments,” said Jessicamarie. “I became a lab manager and purchased scientific supplies, arranged meetings and interviews, and gained so much knowledge from my lab coworkers.”

“I had a great-uncle who died from AIDS in the early 2000s so I feel I can make a difference through working in an HIV research lab,” said Jessicamarie. “And I love the people I work with. Everyone is helpful and supportive.”

Kwasi, like Jessicamarie, graduated from Wistar’s BTT and BRT programs with critical hands-on skills as well as the analytical thinking needed to carry out the right experiments for a research project — knowledge he could only gain through on-the-job experience.

“Every aspect of the apprenticeship program was meaningful to me and applies to the work I’m now doing in the COVID-19 space,” said Kwasi. “But most useful is the intense knowledge I gained from having my own project. This challenged me to be more disciplined: I learned how to follow a plan, respect the schedule I set to carry out the research and leave no stone unturned.”

Wistar’s education and training programs, deeply rooted in the Institute’s mission to train the next generation of scientists, are two workforce development models that successfully create cross-boundary collaborations between industry, academia and research institutes, offer in-depth training and education while providing students a wage, and give trainees the skillsets sought after by the life sciences industry.

Wistar’s broad and deep connections to scientists in academia, biotech, government, and pharma, both regionally and nationally, continue to drive opportunities to train scientists at every level and from all backgrounds.

Spotlight on Wistar COVID-19 Researcher: Paul Lieberman, Ph.D.

Creating a new drug from scratch takes many years and billions of dollars. While discovery of new molecules against SARS-CoV-2 and other emerging viruses is imperative, the world also needs solutions for COVID-19 now. This is why there has been wide interest in repurposing existing drugs, for which safety and pharmacologic profiles are already available, as a viable strategy to save critical time and resources and quickly discover potential candidate drugs to combat the pandemic. 

Dr. Paul Lieberman is a molecular virologist in Wistar’s Cancer Center, better known for his pioneering studies on how certain viruses persist in the body in a latent, long-term infection that can lead to cancer. He leads a talented  drug-discovery team that has created first-in-class molecules directed against  Epstein-Barr virus (EBV) as a novel therapeutic approach for potentially treating EBV-associated cancers including Burkitt’s lymphoma, nasopharyngeal carcinoma and Hodgkin’s lymphoma.

Applying their antiviral drug discovery expertise and innovative methods to “drug” the RNA component of viruses and cancer cells, the Lieberman lab is now working on a drug-repurposing project to quickly identify FDA-approved molecules that trap and inactivate SARS-CoV-2 RNA, the genetic material that carries the life information for the virus, just like DNA does in our cells. 

SARS-CoV-2 RNA forms unique three-dimensional structures known as pseudoknots that are essential for viral replication and for the ability of the SARS-CoV-2 virus to cause disease. RNA pseudoknots may be a promising new target for therapeutic intervention. 

Working closely with the Molecular Screening Facility, which provides state-of-the-art technologies and industry standard expertise, Dr. Lieberman and his team rapidly set up a high-throughput, novel screening assay based on identifying molecules that bind to the SARS-CoV-2 pseudoknot structures and disrupt their function. His team is in the process of evaluating thousands of FDA-approved small molecule drugs. 

The lead candidates identified in the screen will then be further tested in cells and in preclinical models for their ability to stop infection and disease progression, and eventually to advance the best molecules into the clinic.

“We are taking a new, very focused approach to evaluate existing molecules,” said Dr. Lieberman. “This allows us to look for a very specific activity to deploy against the virus in molecules that have already tested safe in humans.”

Because RNA pseudoknot structures are similar in other coronaviruses, the outcome of this project could also have broader applications against other respiratory diseases.

New Research Highlights a Rare Gene Mutation Linked to Cancer Risk in the Ashkenazi Jewish Population

Dr. Maureen Murphy, Ira Brind Professor and program leader of the Molecular & Cellular Oncogenesis Program of The Wistar Institute Cancer Center, has been investigating the importance of inherited mutations in the gene encoding p53 tumor suppressor protein to determine cancer susceptibility in people of African descent and people of Ashkenazi Jewish descent, for the past twenty-two years.

Dr. Murphy published her most recent scientific findings in the journal Cancer Research, together with her research collaborators from more than eleven research hospitals throughout the country, including the University of Pennsylvania and Children’s Hospital of Philadelphia.

The p53 protein is one of the body’s best defenses against cancer and is necessary for response to chemotherapy, overseeing the cell’s fate after assaults that cause DNA damage and other stress stimuli. Controlling several different cellular responses, just like the director of an orchestra keeps all the instrument sections and their musical notes synchronized, p53 decides if cell damage can be repaired or calls for the extreme solution — eliminating the affected cells to prevent malignant transformation. It is through the fundamentally important scientific studies connecting basic science with clinical care, such as Dr. Murphy’s latest publication, that the world is becoming better positioned to implement targeted personalized medicine approaches to improve health care.

Dr. Murphy describes p53 as the most important protein in our body. She explained the significance of the new study and how it came to be in this Q&A.

Q: Since p53 is so important, what happens when it doesn’t work?

A: Mutations in the p53 gene that are acquired during one’s life can compromise the protein’s function and its ability to restrain cancer development. Indeed, p53 is the most mutated gene in cancer. People who are born with inherited p53 mutations have a 90 percent chance of developing cancer in their lifetime, including soft tissue and bone sarcomas, breast cancer and leukemia, a rare condition called Li-Fraumeni Syndrome (LFS). Often, these tumors arise during childhood or early in life, and individuals have multiple tumors throughout their lifetime. These families frequently describe their experience as being “stalked by cancer”.

Q: How important is it for these individuals to know their genetic background?

A: Diagnosis of LFS is critical to help affected families understand their risk and seek appropriate screening for early cancer detection. In these cases, cancer screening is extremely aggressive. Unfortunately, there are many different p53 mutations that correspond to a wide variety of cancer types and risk levels, and we do not yet know if all mutations require the same level of screening. Therefore, it’s very important to identify those mutations and understand what they mean for patients in terms of cancer risk and age of onset.

Q: What did you discover in this new study?

A: Studying eight different families, we identified a rare, lower-risk p53 mutation that is associated with LFS cancer types, but at slightly lower rates and with significantly later age of onset of pediatric cancers. This mutation causes a milder defect in p53 function than other LFS p53 mutations — something we call ‘hypomorphic mutations,’ which result in reduced expression or functional performance of a protein, but not a complete loss. Hypomorphic mutations are rare and typically tend to recur within one ethnicity. We refer to these as ‘founder’ mutations because one or more ancestors in the affected population were carriers of the altered gene and passed it down to their descendants. This specific mutation tracks within the Ashkenazi Jewish population.

Q: How did you start this collaboration with so many other institutions to study this p53 mutation?

A: I gave a seminar at Penn and Dr. Maxwell was in the audience. A few days later, she wrote to me saying she had interesting cancer cases in her clinic and we decided to work together to figure out whether there was a new hypomorphic mutation behind those cases. Science at its best is when we are able to join forces and combine our questions and expertise to solve a biological puzzle and ultimately advance patient treatment.

Q: What pieces of the p53 puzzle are still missing?

A: We need to continue these types of studies to better understand the p53 genetic landscape across populations and the implications of each mutation. I believe that will inform better clinical management of patients and, importantly, help find new specific therapies. We know that people who carry these hypomorphic mutations respond poorly to standard cancer therapy. At the same time, their genetic differences might result in specific vulnerabilities of their tumors. The focus and passion of my lab is to look for these therapeutic vulnerabilities and exploit them to find drugs that can do better for these patients so that we can ultimately tailor therapy to their specific genetic background. We think that a personalized medicine approach, based upon the genetics of the individual and the tumor, is the next frontier for cancer research, and we aim to be at the cutting edge of this frontier.