Skip to main content

Author: The Wistar Institute

Enhanced Anticancer Compound May Allow for Precise Activation and Tracking of Treatment

PHILADELPHIA — (May 15, 2019) — Researchers at The Wistar Institute and the University of South Florida have advanced a novel compound that specifically targets the endoplasmic reticulum (ER) stress response that is frequently hyperactivated in cancer and promotes survival of cancer cells during stressful conditions. The new compound has unique chemical properties that allow for precise activation and can be used for tracking its activity in vivo thanks to the emission of fluorescence. Research was published online in the Journal of Medicinal Chemistry.

The ER is a cellular structure that oversees protein folding and assembly, and it activates stress response mechanisms in response to the accumulation of misfolded proteins or other stressful conditions, including low oxygen.

“Certain cancers rely on the protective role of the ER stress response to sustain their growth in stressful environmental conditions,” said Chih-Chi Andrew Hu, Ph.D., associate professor in Wistar’s Immunology, Microenvironment & Metastasis Program, and co-lead author on the study with Juan R. Del Valle, Ph.D., from the University of South Florida. “We and other groups showed that genetically and pharmacologically targeting the ER is a very effective way to attack various tumors.”

The Hu lab has been actively advancing a research program for development of compounds to block vital functions of the ER stress response, specifically inhibitors of the IRE1 protein that senses the ER stress and responds by activating a cascade of cellular events. In addition to inducing tumor cell apoptosis, these compounds activate antitumor T-cell function and inhibit tumor-associated immunosuppressive cells in vivo in mouse models, resulting in tumor shrinkage.

“This class of inhibitors has shown therapeutic promise in several cancer types, including chronic lymphocytic leukemia and Burkitt lymphoma,” said Hu. “Therefore, we are very interested in advancing them to the clinic.”

In this study, through a series of chemical modifications, Hu and collaborators created a molecule called PC-D-F07 that is pharmacologically inactive and requires UV irradiation to be “turned on” and converted into the active D-F07, which is a very potent inhibitor of IRE-1 function.

The novel prodrug strategy enhances the efficacy of IRE-1 inhibitors and allows fine control of their activity at a precise time and location by UV irradiation. Importantly, once active, D-F07 emits fluorescence that can be tracked in cells and potentially in vivo, providing a real-time therapy readout. This new strategy for stimulus-mediated release of pharmacological activity provides a promising platform to target the ER stress response for anticancer therapy and for developing other potent inhibitors.

Co-authors: First author Andong Shao, Chih-Hang Anthony Tang, Qin Xu and Claire Phoumyvong from Wistar; Chang Won Kang, Christopher Cain and Juan R. Del Valle from University of South Florida.

Work supported by: National Institutes of Health (NIH) grants R01CA163910, R21CA199553, and R01CA190860.

Publication information: Structural tailoring of a novel fluorescent IRE-1 RNase inhibitor to precisely control its activity, Journal of Medicinal Chemistry (2019). Advance 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.org.

A Tale of Families, Genes and Resilience

Recently, Wistar had the great honor of bestowing its annual Women & Science’s Helen Dean King Award upon Dr. Mary-Claire King, one of the most esteemed and productive scientists of our time, in recognition of her seminal contributions to science and human health.

King needs no introduction as the impact of her work in human genetics speaks volumes: She established the hereditary nature of breast cancer and identified mutations in the BRCA1 gene as the culprit of the phenomenon, shifting the view of the role of genes in cancer.

She also put her expertise at the service of humanitarian efforts around the world, applying genomic sequencing to identify victims of human rights abuses by, for example, reuniting more than 100 children who were kidnapped during the “Dirty War” in Argentina with their biological families.

After the award ceremony, with extreme clarity and verve King presented a fascinating overview of how an evolutionary biologist by training, she took on the task of deciphering the mystery of breast cancer heredity, from the first observations in the pre-genetics era through the Human Genome Project revolution, to the functional studies of the BRCA1 protein and the current options for prevention and targeted therapy.

King traced the beginning of the story back to the 1860s, when French surgeon and pathologist Paul Broca described familial breast cancer for the first time in medical history and postulated the existence of “unknown dispositions of the body which precede the first appearance of certain tumors.” Boca didn’t know genetics but based his conclusions on the study of family trees and autopsy cancer tissues. Later, in the 1970s, Broca’s hypothesis was formalized with statistical and epidemiological studies showing that women with a first-degree relative affected by breast and ovarian cancer had a higher risk of developing the disease.

This is when King entered the field of cancer epidemiology and genetics and began her “hunt” for what turned out to be the genetic origin of breast cancer. At that time, cancer research mainly focused on the role of viruses as the cause of cancer, so King’s working hypothesis of a genetic transmission of breast cancer was regarded with strong skepticism. Thanks to her background in evolutionary biology, she approached the question by building mathematical and statistical models to search for parameters that could explain the patterns of breast cancer in some families. She became convinced that the alteration of a single gene was the answer.

As King recalls, a lucky coincidence happened in the early 80s when the National Cancer Institute (NCI) was conducting a large epidemiological study to analyze the relationship between the use of oral contraceptives and breast cancer. King asked Dr. Frank Rauscher Jr., then NCI director, to include a question to study participants regarding family history of breast cancer. This resulted in data from more than 1,500 women, which King compared with her mathematical predictions, confirming her model of genetic transmission of breast cancer risk and existence of a breast cancer gene.

This model postulated that women and men in high-risk families inherit mutations in one of their two copies of this gene in all the cells of their body. These individuals remain healthy until a second mutation, referred to as “second hit,” alters the other copy in breast and ovarian cells, completely removing the gene function in these tissues. This pattern of transmission, known to geneticists as autosomal dominant, explains why mutation carriers have a much higher risk of cancer and, at the same time, why cancer doesn’t occur in 100% of the cases, requiring a second mutation event.

A few years later, in the mid 90s, similar studies conducted on the Eastern European Ashkenazi Jewish population confirmed King’s results on a completely different set of families, showing that women carrying mutations in breast and ovarian cancer susceptibility genes had a 70-80% risk of developing breast cancer, the same King estimated in the 70s with her mathematical model.

In the late 80s, however, the existence of a breast cancer gene was still a theory. To prove it, King had to find it or, as she said, “hold the gene in her hands.” Without the molecular biology technologies available today, it took a total of 17 years of hard and diligent work to finally hunt down the gene, later named BRCA1 and map it to chromosome 17. The results of this long effort, published in 1990 in Science, initiated a race to sequence BRCA1, eventually won by Myriad Genetics in 1994.

Soon after, another gene named BRCA2 was discovered, whose mutations are transmitted in the same pattern as BRCA1. It only took one year to sequence the full BRCA2 gene because at that point the Human Genome Project was underway and thanks to new technologies that had become available.

Thanks to King’s pioneering work, it is now well established that women who carry mutations in BRCA1 have more than 60% and close to 40% risk of developing breast and ovarian cancer, respectively, before age 70. For carriers of a BRCA2 mutation, the risk is around 45% for breast and 25% for ovarian cancer. More breast cancer genes have also been identified.

King is a fierce advocate for prevention in at-risk women and those in the general population, as in some cases, when family history is not available or the mutation is inherited from fathers who didn’t develop cancer, identifying women at risk can be challenging.

In a Journal of the American Medical Association (JAMA) commentary in 2014, King maintained that every woman should be offered genetic screening of breast cancer susceptibility genes by age 30 as a routine practice, so that carriers can opt to undergo risk-reducing surgery to remove their breasts and ovaries or remain under strict surveillance for early cancer detection.

“Every BRCA mutation detected after diagnosis is a missed opportunity to prevent cancer,” said King in closing her talk. “No women should die of breast or ovarian cancer, because these diseases can be preventable with the right screening approach.”

2019 Helen Dean King Award Ceremony

Wistar Joins in a Celebration of Science Education at the Philadelphia Science Festival

Along with warmer temperatures and the beautiful blossoming across the city, spring has become synonymous with the Philadelphia Science Festival, a staple and model for similar initiatives in other cities, that continues to grow each year.

Established in 2011 by The Franklin Institute as one of the first of its kind in the country, the Festival is a celebration of science, technology and education in everyday places — parks, libraries, museums, restaurants, and bars. More than 200 partners, including the major academic, research and educational institutions in the area, collaborate to produce a nine-day series of science, technology, engineering, and mathematics (STEM)-focused events for adults and children alike.  

The Festival aims to engage the community and inspire the next generation of scientists and engineers by creating homegrown citizen scientists. 

Wistar is proud to be a partner and to have served on the advisory council for the Festival from the beginning. This year, a team of Wistar volunteers will participate in Festival events with hands-on, educational activities designed to inform the public about basic cancer and infectious diseases and spread awareness about Wistar science. Wistar’s commitment to the Science Festival aligns with its mission of educating new generations of scientists and engaging the community in its work to improve human health. 

The Festival culminates in the free, daylong Science Carnival on the Ben Franklin Parkway on May 4 from 10:00 a.m. until 4:00 p.m. A group of Wistar scientists, trainees and staff will share their passion for science with Festival attendees at the Wistar booth in zone 6/Blue. Volunteers work hard each year to explain vaccine development or how a cell functions, but they are rewarded by the enthusiasm and curiosity of those that visit the Wistar tent. 

For more info and full Festival program, visit www.fi.edu/psf.

How CURE Boosts the Health of Pennsylvanians

Each year in early May, representatives from Wistar and fellow institutions who are part of the Pennsylvania Cancer Alliance engage legislators to reinforce the importance and impact of CURE funding.

The Commonwealth Universal Research Enhancement Program (CURE), which funds health research across Pennsylvania, was established in 2001 with money received by the state from the Tobacco Master Settlement Agreement in 1998.

This was the largest civil settlement in U.S. history and came as the result of a lengthy legal battle to get tobacco companies to admit to the well-established causal relation between cigarette smoke and cancer and other health conditions.

The settlement was historic and far-reaching. Tobacco companies agreed to pay $206 billion to 46 states and the District of Columbia over the first 25 years. They also agreed to end marketing to kids and to provide annual payments indefinitely to cover some of the healthcare costs of smoking-related illnesses.

The states involved in the settlement agreed to devote their payments to reduce tobacco use. Pennsylvania is one of the few states that have invested 100% of settlement proceeds in health-related enterprises.

A few years after the settlement, then-governor Tom Ridge signed the Tobacco Settlement Act into law. The act allowed the Department of Health (DOH) to use settlement money to establish the CURE program with the goal of supporting research that can improve the health of Pennsylvanians.

CURE provides funding to universities, hospitals and nonprofits for broad-based health research, tobacco use prevention and cessation programs, and hospital uncompensated care programs. There are two types of grants:

  • Formula — Institutions that receive funding from the National Institutes of Health or the National Cancer Institute (NCI) are eligible for these non-competitive grants.
  • Non-formula — These competitive grants are distributed by the DOH, which each year assigns a priority for these grants. The 2018-2019 priority is collaborative research on opioid abuse and the overdose crisis.

Through the CURE Program, 39 organizations across the state, including Wistar, have received funding, and more than 85 patents have been filed for commercial use because of CURE-sponsored research. The settlement money has also generated more than 1,000 healthcare jobs in Pennsylvania.

Sustainable funding for CURE is essential because not only does it improve the economy, but it also provides Pennsylvania citizens with significant health benefits through research and health advances.

CURE Funding at Work at Wistar

At The Wistar Institute, pilot CURE funds were essential for Dr. Louise Showe, a preeminent researcher focused on identifying novel predictive cancer biomarkers, to advance research that has subsequently been translated into a new minimally invasive blood test that will help clinicians to distinguish benign lung nodules from cancerous lung nodules in high-risk populations. This test, developed with a for-profit research and development partner, is expected to be commercially available soon. Without CURE funding, Showe’s research would have taken much longer to complete and with limited preliminary data, it would have been impossible to attract a partner to assist with the further development and commercialization of a diagnostic test.

Wistar also uses CURE money to underwrite research by junior investigators at the start of their independent research careers. National Institutes of Health (NIH) grants are highly competitive and are generally not awarded to investigators until they have significant progress in their chosen area of inquiry. As a consequence, institutions must find support for their young investigators until they become competitive for NIH funding.

In addition to jump-starting biomedical research careers, CURE money is used to accelerate the translation of new discoveries into future potential products that may change the way cancer and infectious diseases are diagnosed and treated. For example, Drs. Alessandro Gardini, Kavitha Sarma, and Jessie Villaneuva have been able to elucidate novel mechanisms underlying tumor initiation and treatment resistance in ovarian cancer, glioblastoma and melanoma, respectively, with the support of CURE funding. Drs. Luis Montaner, Zachary Schug, Joseph Salvino, and Farokh Dotiwala have applied CURE funds towards the research and discovery of novel small molecule inhibitors useful for treating ovarian, colorectal cancers, gliomas and antibiotic-resistant microorganisms including tuberculosis, respectively.

CURE funds are critical for seeding transformative discoveries that can lead to the next generation of effective therapies.

Wistar Attends 2019 CURE Roundtable at the Pennsylvania State Capitol in Harrisburg

Wistar Earns ‘Exceptional’ Rating for NCI Cancer Center Support Grant for the Second Review Cycle in a Row, Demonstrating Strength of Science 

PHILADELPHIA — (April 23, 2019) — After extensive peer review, The Wistar Institute’s Cancer Center has received an “exceptional” rating on its Cancer Center Support Grant (CCSG) application from the National Cancer Institute (NCI) for the second review cycle in a row and the grant has been recommended for renewal, providing $13.6 million to the Cancer Center scientific enterprise.

Wistar’s CCSG achievement of “exceptional” denotes its prestigious standing among the nation’s best, a status gained and maintained through top research programs, historic partnerships, state-of-the-art core facilities, impactful educational programs, and scientific leadership.

“This recognition is an incredible accomplishment for Wistar and a testament to the scientists, staff and collaborators who continue to shape and drive our cutting-edge science,” said Dario C. Altieri, M.D., president and CEO, director of the Cancer Center and the Robert and Penny Fox Distinguished Professor at The Wistar Institute. “We have a storied history of producing high-impact scientific discoveries in cancer biology and through this NCI Support Grant will be able to continue to advance our influence on translational and patient-focused cancer research.”

In 1972, Wistar became the first institution in the nation to receive the NCI-designation as a cancer center solely devoted to basic research in cancer biology. Since then, the Institute has continuously held this designation, achieving the highest ranking of “exceptional” for its CCSG renewal application.

To earn the NCI designation, Wistar’s Cancer Center demonstrated scientific excellence, innovative and impactful contributions in basic and translational cancer research, and productive partnerships with clinical collaborators and neighboring academic institutions. The center’s activities were evaluated in a rigorous peer review process. Key accomplishments that fostered scientific achievements during this process include:

  • Expanded scientific infrastructure including the opening of the Robert and Penny Fox Tower;
  • Transition of leadership to Dario C. Altieri, M.D., Wistar President and CEO;
  • Expanded five-year strategic plan;
  • Successful recruitment of 11 new talented Cancer Center principal investigators; and,
  • Expansion of the collaboration with the Helen F. Graham Cancer Center & Research Institute.

The Wistar Institute Cancer Center’s overall mission is to advance and merge basic, translational and clinical cancer research in a single scientific continuum, and to do so by pursuing scientific excellence, innovative contributions and productive collaborations with diverse academic institutions.

Additionally, Wistar’s many partnerships locally and beyond have enabled the Institute to move research forward for the benefit of human health. In partnership with the Helen F. Graham Cancer Center in Delaware, one of the largest community cancer hospitals in the nation, Wistar scientists can access critical clinical cancer related data and patient samples to drive discovery research that may someday translate into new therapies. The Institute’s 20+ year synergetic educational collaboration with the Community College of Philadelphia has created innovative training opportunities to foster a pipeline of young scientists equipped and eager to enter the region’s vibrant life sciences community.

“Through our partnerships and state-of-the-art facilities, we continue to drive innovative research forward, ignite productive collaborations, publish our science in high-impact journals, recruit top scientists in the field, and offer outstanding facilities and technical expertise to our scientists,” said Rugang Zhang, Ph.D., deputy director of Wistar’s Cancer Center and professor and co-leader of the Gene Expression & Regulation Program at Wistar. “This NCI designation of our Cancer Center recognizes Wistar’s outstanding research efforts, and our deep commitment to advancing cancer research and care.”

###

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.

Wistar Science Highlights: Zika Infection, Cancer Immunology and Immunotherapy

In a study published online in Nature, the lab of Dmitry I. Gabrilovich, M.D., Ph.D., Christopher M. Davis Professor and leader of the Immunology, Microenvironment and Metastasis Program, identified a critical step that happens when parts of the immune system switch sides and work for the tumor.

Myeloid derived suppressive cells (MDSCs) are a bone marrow-derived cell population that becomes pathologically activated in cancer patients and hinders the body’s antitumor immune response. They are associated with poor prognosis and resistance to immunotherapy.

“We need to better define how these cells work and what pathways are important, so that we can devise strategies to keep them at bay,” said Gabrilovich. “Our study represents an important step forward in that direction.”

They found that a gene that encodes for a protein called FATP2 is expressed at much higher levels in MDSCs isolated from tumor-bearing mice than in the corresponding cell population derived from healthy mice. They also described how FATP2 converts normal neutrophils into immunosuppressive cells in the presence of a tumor.

This study opens new opportunities for specific therapeutic targeting of MDSCs to inhibit tumor growth and enhance the anticancer activity of other immunotherapies. READ MORE


David B. Weiner, Ph.D., executive vice president and director of Wistar’s Vaccine & Immunotherapy Center, and W.W. Smith Charitable Trust Professor in Cancer Research, and his collaborators described a new approach for delivery of DNA-encoded monoclonal antibodies (DMAbs) for protection against Zika virus (ZIKV) infection.

In this study, published in Molecular Therapy, the lab engineered a synthetic DNA plasmid encoding a previously identified potent anti-ZIKV monoclonal antibody. When injected intramuscularly in mice and non-human primates, these DMAbs resulted in antibody presence in circulating blood for several weeks to months and provided long-term protection against infection in both small animals and, for the first time, non-human primate preclinical models.

“These properties, coupled with the ease of production and storage, support further development of DMAbs as a possibly ideal approach during infectious disease outbreaks to provide rapid protection to at-risk individuals and, in the case of Zika, to their offspring as well,” said Weiner. READ MORE


A study by researchers in Wistar’s Vaccine & Immunotherapy Center, published online in JCI Insights, led to the creation of a novel synthetic DNA approach for patient-specific production of cancer-targeting molecules called bispecific T cell engagers (BiTEs).

BiTEs are a type of artificial monoclonal antibody that directly binds to tumor targets, while at the same time binding to and activating killer T cells. In doing so, BiTEs act as a bridge between killer T cells and tumor cells, making sure that T cells see and specifically kill tumors with high accuracy.

BiTE production is difficult and they have a short duration in the bloodstream, requiring patients to be continuously infused for up to seven days for the treatment to be effective.

Teams led by Weiner and Kar Muthumani, Ph.D., assistant professor in the Vaccine & Immunotherapy Center, developed DNA-encoded bispecific T cell engagers (dBiTEs) that, when injected into the muscle in mouse models, provided the genetic instruction for muscle cells to make and launch the novel molecule directly into the bloodstream to seek and destroy tumors.

They designed dBiTEs specific for HER2, a cancer antigen found on breast and ovarian cancer cells, and tested them in mouse models of ovarian cancer. HER2 dBiTEs were highly expressed after just a single injection and were able to attract T cells that were activated to kill the tumor cells. Importantly, the tumor-killing activity persisted for more than a month.

“These results showed that dBiTEs are much more potent than traditional monoclonal antibodies. Not only did treatment extend survival of tumor-bearing mice, but also 80% of the animals treated with dBiTE were cured – a high bar to reach in this animal model,” said Muthumani. “Our results show that further exploration of the dBiTE approach for therapeutic development is warranted.” READ MORE

Signed, Sealed and Credentialed: Biomedical Technician Training Program is an Accredited Pre-apprenticeship Program in the Commonwealth of PA

For two decades, Wistar’s Biomedical Technician Training (BTT) pre-apprenticeship program has injected biomedical talent into the region’s life sciences workforce by ushering class after class of promising Community College of Philadelphia (CCP) students to attain the skills and experience required to become part of a growing biomedical science workforce. Director of the Pennsylvania Apprenticeship and Training Office Eric Ramsey and the PA Department of Labor & Industry officially recognized the BTT Program as a state-credentialed, non-traditional, biomedical pre-apprenticeship program.

“Across the nation, states look to strengthen their pipeline of skilled workers and on-the-job training opportunities to keep talent and jobs local,” said Brian Keith, Ph.D., Wistar dean of biomedical studies. “Within Pennsylvania, Wistar’s BTT Program has been providing students with highly competitive skills that prepare them for success as research assistants in the burgeoning life sciences workforce.”

Through learn-and-earn classwork and on-the-job laboratory training, CCP students from diverse backgrounds embark on an alternative to the traditional coursework track. Taught by experienced mentor-scientists, they learn biomedical techniques and skills while bringing out-of-the-box problem solving skills and a strong work ethic from diverse perspectives to enhance the STEM/biomedical workforce.

“What started as a commitment to developing a pipeline of research lab technicians for the burgeoning life sciences region in Philadelphia has become much more than the sum of its parts,” said William Wunner, Ph.D., Wistar director of outreach education, technology training and academic affairs. “In the BTT Program, I see the growth potential of our students—some are looking to break into the life sciences industry and some are setting their sights on attaining further education degrees and pushing beyond. Either choice makes the region stronger.”

Innovative Synthetic DNA-based Cancer Immunotherapy Approach Shows Tumor-clearing Ability in Preclinical Models

PHILADELPHIA — (April 18, 2019) — Wistar scientists have developed a novel synthetic DNA approach for patient-specific production of cancer-targeting molecules called bispecific T cell engagers. DNA-encoded bispecific T cell engagers (dBiTEs) designed against the HER2 protein were tested in preclinical models of ovarian cancer and induced tumor regression, demonstrating the potential of this novel approach for immunotherapy. Study results were published online in JCI Insights.

BiTEs are a type of artificial monoclonal antibody that directly bind to tumor targets, while at the same time binding to and activating killer T cells, providing these T cells with the ability to see and specifically kill the tumors with high accuracy. BiTEs were developed as biologically produced molecules. The first BiTE was recently approved for treatment of a form of B-cell cancer.

While there is a great deal of interest in this type of approach, BiTEs development has been slow, in part due to difficulty of production and their short bloodstream half-life of just a few hours, according to lead researcher David B. Weiner, Ph.D., executive vice president and director of Wistar’s Vaccine & Immunotherapy Center, and W.W. Smith Charitable Trust Professor in Cancer Research. Accordingly, for the currently approved BiTE, patients are required to be continuously infused for up to seven days for the treatment to be effective. A simpler, more cost-effective way to provide patients with such important molecules through a technology that would also allow for rapid design modification would be an advantage.

“Leveraging our expertise in designing synthetic DNA molecules that can be produced in vivo and building on the DNA-encoded monoclonal antibody technology that Wistar’s Vaccine & Immunotherapy Center has been advancing, we developed the dBiTE approach that could make these molecules available for treating more patients,” said Weiner. “In our preclinical studies, dBiTEs demonstrated a unique profile compared to conventional BiTEs, overcoming some of the technical challenges associated with production and delivery of BiTEs and permitting combinations of dBiTEs to be administered at one time as a multi-pronged approach to treat resistant cancer.”

Wistar and colleagues at Inovio Pharmaceuticals, Inc., focused on advancing new synthetic DNA designs for BiTE-like molecules by engineering and encoding them in optimized synthetic plasmid DNA cassettes. dBiTEs are then injected locally into the muscle and muscle cells convert the genetic instructions into protein to allow for direct in vivo launching of the novel molecule directly into the bloodstream to the seek and destroy tumors.

Through reengineering a DNA-encoded monoclonal antibody (DMab) that the lab had previously designed to target HER2, an important cancer antigen found on breast and ovarian cancer cells, the team designed HER2-specific dBiTEs. Tested in mouse models of ovarian cancer, HER2 dBiTEs were highly expressed after just a single injection and were able to bind to their molecular target on the tumor and attract T cells that were activated to kill the tumor cells. Importantly, the tumor killing activity persisted for more than a month.

“These results showed that dBiTEs are much more potent than traditional monoclonal antibodies. Not only did treatment extend survival of tumor-bearing mice, but also 80% of the animals treated with dBiTE were cured – a high bar to reach in this animal model,” said co-lead researcher Kar Muthumani, Ph.D., assistant professor in the Vaccine & Immunotherapy Center at Wistar. “Our results show that further exploration of the dBiTE approach for therapeutic development is warranted.”

Co-authors: First author Alfredo Perales Puchalt, Elizabeth K. Duperret, Xue Yang, Patricia Hernandez, Krzysztof Wojtak, Xizhou Zhu, Seang-Hwan Jung, Edgar Tello-Ruiz, and Luis J. Montaner from Wistar; Megan C. Wise from Inovio.

Work supported by: National Institutes of Health (NIH) Special Program of Research Excellence grant P50 CA174523 to Wistar and the University of Pennsylvania, and NIH grants T32-AI055400 and F32 CA213795. Additional funding was provided by the W.W. Smith Charitable Trust, the Basser Foundation and Inovio. Core support for Wistar was provided by Cancer Center Support Grant P30CA010815.

Publication information: DNA-encoded bispecific T cell engagers and antibodies present long-term
antitumor activity, JCI Insight (2019). Advanced 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.

Identification of a Critical Regulator of Immune Suppressive Cells Reveals a Novel Target for Cancer Immunotherapy

PHILADELPHIA — (April 17, 2019) — Wistar scientists have identified a critical regulator of the function of myeloid derived suppressive cells (MDSCs), opening new opportunities for specific therapeutic targeting of these cells to inhibit tumor growth and enhance the anticancer activity of other immunotherapies. Study results were published online in Nature.

MDSCs are a bone marrow-derived cell population that becomes pathologically activated in cancer patients and inhibits the body’s antitumor immune response. They are associated with poor prognosis and resistance to immunotherapy. The mechanisms responsible for conversion of these cells into immune suppressive cells are not well defined.

“MDSCs have emerged as fundamental players in the fight of our immune system against cancer and the success of certain therapies,” said senior author Dmitry I. Gabrilovich, M.D., Ph.D., Christopher M. Davis Professor and program leader of the Immunology, Microenvironment and Metastasis Program at Wistar. “We need to better define how these cells work and what pathways are important, so that we can devise strategies to keep them at bay. Our study represents an important step forward in that direction.”

Gabrilovich and collaborators delved into MDSC lipid metabolism because the accumulation of lipids has been shown in several immune cells in the tumor microenvironment. They found a specific role for a protein called FATP2, which mediates the intake of fatty acids.

They compared MDSCs isolated from mice carrying different tumor types with the corresponding cell population in healthy mice and observed a much higher expression of the gene that encodes for FATP2, specifically in polymorphonuclear MDSCs (PMN-MDSCs), which are the immunosuppressive counterpart of neutrophils. They showed that FATP2 mediates the immunosuppressive activity of PMN-MDSCs.

At the molecular level, the team found that FATP2 selectively allows for the accumulation of a type of fatty acid called arachidonic acid, which is needed for the cells to make prostaglandin E2 (PGE2), a potent inflammatory molecule that is also implicated in the immunosuppressive activity of MDSCs in cancer. They also characterized the signaling pathway that activates transcription of the FATP2 gene in PMN-MDSCs.

Importantly, the observations obtained in the mouse models were recapitulated in PMN-MDSCs isolated from cancer patients. These cells have a higher lipid content and higher expression of FATP2 than the counterpart isolated from healthy donors.

“The obvious next step in our research was to verify whether FATP2 was a good candidate from a therapeutic standpoint,” said Filippo Veglia, Ph.D., a research assistant professor in the Gabrilovich Lab and first author on the study. “We were excited to find that a selective inhibitor of FATP2 called lipofermata caused a significant delay of tumor growth in mice.”

Researchers confirmed this effect was specifically due to activation of the immune system, as it was not observed in immunodeficient mice and after depletion of CD8+ T cells, the cells in charge of destroying the tumor. The antitumor effect was even stronger when lipofermata was given in combination with other immunotherapeutic agents — while the single treatments slowed down tumor progression, most of the mice that received the combined treatment completely rejected the tumors.
Gabrilovich and colleagues identified a critical regulator of the function of PMN-MDSCs and described pharmacological inhibition of FATP2 as a novel and specific therapeutic strategy to block the immunosuppressive activity of these cells and enhance the effect of cancer therapy.

Co-authors: Alessandra De Leo, Andrew Kossenkov, Laxminarasimha Donthireddy, Zach Schug, Subhasree Basu, Fang Wang, Maureen E. Murphy, Paul M. Lieberman, Cindy Lin, and Yulia Nefedova from Wistar; Vladimir A. Tyurin and Valerian E. Kagan from the University of Pittsburgh; Maria Blasi from Duke University School of Medicine; Tsun Ki Jerrick To, Emanuela Ricciotti, Gregory Masters, and Robert H. Vonderheide from University of Pennsylvania School of Medicine; Concetta DiRusso and Paul Black from University of Nebraska; Charles Mulligan, Brian Nam, Neil Hockstein, and Michael Guarino from Helen F Graham Cancer Center at Christiana Care Health System.

Work supported by: National Institutes of Health (NIH) grants CA R01CA165065 and AI110485. Core support for Wistar was provided by Cancer Center Support Grant P30CA010815.

Publication information: Fatty acid transporter 2 reprograms neutrophils in cancer, Nature (2019). Advance 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.org.

Synthetic DNA-encoded Antibodies Against Zika Virus Shown to be Effective in Preclinical Studies

PHILADELPHIA — (Apr. 5, 2019) — A new approach for delivery of DNA-encoded monoclonal antibodies (DMAbs) has been reported by Wistar scientists and their collaborators. This new technology allows direct production of monoclonal antibody-like molecules in living animals.

In this study, published in Molecular Therapy, the researchers focused on developing novel DMAbs for protection against Zika virus (ZIKV) infection. DMAbs achieved persisting antibody expression that provided long-term protection against lethal virus challenge in both small animal and, for the first time, non-human primate preclinical models. This study provides a direct bridge to the clinic, supporting further development of the new DMAb technology towards wider application.

“We showed that the DMAb platform produces fast and transient but sustained antibody expression in the blood of small and large animal models,” said lead researcher David B. Weiner, Ph.D., executive vice president and director of Wistar’s Vaccine & Immunotherapy Center, and W.W. Smith Charitable Trust Professor in Cancer Research. “These properties, coupled with the ease of production and storage, support further development of DMAbs as a possibly ideal approach during infectious disease outbreaks to provide rapid protection to at-risk individuals and, in the case of Zika, to their offspring as well.”

Monoclonal antibodies (MAb) are a major category of therapeutic agents with potential for prevention and treatment of a host of infectious diseases. However, development and delivery of MAbs is expensive and limits its applications. The current study advances the novel DMAb technology by showing expression and protection from an infectious challenge.

ZIKV is a mosquito-borne infection endemic to several areas of the world and has become an important global public health concern, with more than two billion people at risk. No approved vaccine or treatment is currently available for ZIKV infection, which is associated with severe birth defects and neurological complications in adults. Survivors develop ZIKV-specific protective antibodies that are being studied as candidates for development of recombinant monoclonal antibodies for preventive use. However, this approach poses challenges related to development, delivery, manufacturing and storage.

The Weiner Lab engineered a synthetic plasmid DNA encoding an identified and developed potent anti-ZIKV monoclonal antibody ZK190 that binds to the virus envelope. When injected as DMAb intramuscularly in mice and non-human primates, DMAb-ZK190 resulted in antibody presence in circulating blood for several weeks to months.

Importantly, when the animals were challenged with a lethal dose of ZIKV, DMAb-ZK190 provided protection from both infection and disease.

“Our study represents the first evidence of protection with a nucleic acid-encoded antibody in a non-human primate model of infection with any infectious agent,” said Ami Patel, Ph.D., first author on the study and a research assistant professor in the Wistar Vaccine and Immunotherapy Center. “This takes us one step closer to clinical development of the DMAb platform for its deployment in the areas where it is most needed.”

Co-authors: co-first author Rianne N. Esquivel, Sagar B. Kudchodkar, Daniel H. Park, Hyeree Choi, Piyush Borole, Kanika Asija, Mamadou Bah, Shareef Shaheen, and Kar Muthumani from Wistar; Karin Stettler and Davide Corti from Humabs BioMed, SA; Jeff Allen, Janess Mendoza, Stephanie Ramos, Jing Chen, Jian Yan, Trevor R.F. Smith, Kate Broderick, Ghiabe Guibinga, and Laurent Humeau from Inovio Pharmaceuticals; Amy C. Durham from the University of Pennsylvania School of Veterinary Medicine.

Work supported by: National Institutes of Health grant T32-AI055400 and a grant from the Bill & Melinda Gates Foundation.

Publication information: In vivo delivery of a DNA-encoded monoclonal antibody (DMAb) protects non-human primates against Zika virus, Molecular Therapy (2019). Advanced 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.