Skip to main content

Author: The Wistar Institute

Daniel Kulp, Ph.D.

Associate Professor, Vaccine & Immunotherapy Center

Kulp has more than 15 years of experience developing molecular design software and leading protein engineering projects. He joined Wistar from The Scripps Research Institute and International AIDS Vaccine Initiative where he was a principal scientist.

Kulp received a bachelor’s degree in Computer Science and Molecular Biology & Biochemistry from Rutgers, The State University of New Jersey, followed by a Ph.D. in Biochemistry and Molecular Biophysics from the University of Pennsylvania. He completed postdoctoral training in structure-based and experimental protein engineering at Los Alamos National Laboratory.

View Publications

The Kulp Laboratory

215-898-6587

dwkulp@wistar.org

The Kulp Laboratory

The Kulp laboratory focuses on rational vaccine and therapeutic antibody design for a variety of priority infectious diseases (e.g. Lassa Virus, HIV, Influenza) and cancer targets. The ultimate test of the lab’s understanding of B cell immune responses is to design new immunogens that drive predictable antibody maturation. To that end, the lab is interested in the development and application of protein engineering methods for modifying antigen/cell receptor interfaces, antigen/antibody interfaces, antigen surface properties and core stabilization.

Staff
  • Postdoctoral Fellows

    Jinwei Huang, Ph.D.
    Kylie Konrath, Ph.D.

  • Graduate Students

    Michaela Helble (Ph.D.)
    Rumi Habib (Ph.D.)
    Niklas Laenger (Ph.D.)
    Shahlo Solieva (Ph.D.)
    Yuanhan Wu (Ph.D)
    Colby Agostino (Ph.D.)
    Martina Tomirotti (Ph.D.)
    Sam Garfinkle (M.D./Ph.D.)
    Andrew Nelson (M.D./Ph.D.)
    Sarah Kim (M.S.)

  • Research Assistants

    Kelly Bayruns
    Amber Kim
    Joyce Park
    Madison McCanna


Lab Alumni

Susanne Walker, Ph.D. – Merck
Neethu Chokkalingkam – Spark Therapeutics
Sinja Kriete – Lake Erie College of Osteopathic Medicine (D.O. Student)
Nicholas Shupin – Robert Wood Johnson Medical School (M.D. student)
Alana Huynh – University of Rochester (Ph.D. student)

Kulp Lab in the News

  • Wistar Scientists Successfully Engineer a Goldilocks Construct: Therapeutic Antibody Could Be a Future Medicine to Improve Outcomes for Melanoma

    Featured News

  • Highlighting Vaccine Research at The Wistar Institute Through the Penn-CHOP-Wistar Vaccine Symposium

    Featured News

  • Novel Nanoparticle SARS-CoV-2 Vaccine Combines Immune Focusing and Self-assembling Nanoparticles to Elicit More Potent Protection

    Press Release

Selected Publications

View Additional Publications

Continue reading

Andrew Kossenkov, Ph.D.

  • Assistant Professor, Vaccine & Immunotherapy Center

  • Genome Regulation and Cell Signaling Program, Ellen and Ronald Caplan Cancer Center

  • Scientific Director, Bioinformatics Facility 

Kossenkov applies computational approaches to the analysis of various kinds of biomedical high-throughput data in effort to interpret results and visualize complex data.

He obtained his bachelor’s and master’s degrees in computer science and bioinformatics from Moscow Engineering Physics Institute in Russia, and his Ph.D. in biomedical science from Drexel University in Philadelphia. In 2007, Kossenkov joined The Wistar Institute as a postdoctoral fellow in the Showe lab and later became managing director of the Bioinformatics Facility. He was appointed assistant professor in 2019.

View Publications

The Kossenkov Laboratory

215-495-6898

akossenkov@wistar.org

The Kossenkov Laboratory

The Kossenkov laboratory collaborates with several other Wistar labs on research projects in cancer, infectious diseases and basic biological questions, including regeneration and stem cell biology, mitochondrial proteins, transcriptional regulation, synapse biology, blood development, and global epigenetic gene silencing. The lab uses bioinformatics, biostatistics and computational biology approaches in a variety of models. They rely heavily on bioinformatics analysis of drug screen results, next-generation sequencing data and proteomics.

Research

Through a network of internal and external collaborations, the Kossenkov lab provides bioinformatics expertise for high throughput data support, analysis and annotation and complex results visualization. Thanks to Kossenkov’s experience in development of database-oriented gene annotation pipelines and algorithms to visualize highly specific and intricate results, the lab provides flexible and customizable support for a wide range of experiments.

Research collaborations outside of Wistar include teams at Fox Chase Cancer Center, The Children’s Hospital of Philadelphia, the University of Pennsylvania, Drexel University, New York University, Weill Cornell, Harvard, and others.

Kossenkov Lab in the News

Selected Publications

  • A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT

    Kossenkov, A.V., Qureshi, R., Showe, L.C., et al. “A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT”. Cancer Res. 2019 Jan 1;79(1):263-273. doi: 10.1158/0008-5472.CAN-18-2032. Epub 2018 Nov 28.

  • Unique pattern of neutrophil migration and function during tumor progression.

    Patel, S., Fu, S., Mastio,  J., Dominguez, G.A., Purohit, A., Kossenkov, A., Lin, C., Alicea-Torres, K., Sehgal, M., Nefedova, Y., Zhou, J., Languino, L.R., Clendenin, C., Vonderheide, R.H., Mulligan, C., Nam, B., Hockstein, N., Masters, G., Guarino, M., Schug, Z.T., Altieri, D.C., Gabrilovich, D.I. “Unique pattern of neutrophil migration and function during tumor progression.” Nat Immunol. 2018 Nov;19(11):1236-1247. doi: 10.1038/s41590-018-0229-5. Epub 2018 Oct 15

  • IRE1α-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity.

    Song, M., Sandoval, T.A., Chae, C.S., Chopra, S., Tan, C., Rutkowski, M.R., Raundhal, M., Chaurio, R.A., Payne, K.K., Konrad, C., Bettigole, S.E., Shin, H.R., Crowley, M.J.P., Cerliani, J.P., Kossenkov, A.V., Motorykin, I., Zhang, S., Manfredi, G., Zamarin, D., Holcomb, K., Rodriguez, P.C., Rabinovich, G.A., Conejo-Garcia, J.R., Glimcher, L.H., Cubillos-Ruiz, J.R. “IRE1α-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity.” Nature. 2018 Oct;562(7727):423-428. doi: 10.1038/s41586-018-0597-x. Epub 2018 Oct 10.

  • CARM1-expressing ovarian cancer depends on the histone methyltransferase EZH2 activity.

    Karakashev, S., Zhu, H., Wu, S., Yokoyama, Y., Bitler, B.G., Park, P.H., Lee, J.H., Kossenkov, A.V., Gaonkar, K.S., Yan, H., Drapkin, R., Conejo-Garcia, J.R., Speicher, D.W., Ordog, T., Zhang, R. “CARM1-expressing ovarian cancer depends on the histone methyltransferase EZH2 activity.” Nat Commun. 2018 Feb 12;9(1):631. doi: 10.1038/s41467-018-03031-3.

  • RNA-Seq of Kaposi’s sarcoma reveals alterations in glucose and lipid metabolism.

    Tso, F.Y., Kossenkov, A.V., Wood, C., et al. “RNA-Seq of Kaposi’s sarcoma reveals alterations in glucose and lipid metabolism.” PLoS Pathog. 2018 Jan 19;14(1):e1006844. doi: 10.1371/journal.ppat.1006844. eCollection 2018 Jan.

View Additional Publications

Continue reading

With Support from the National Science Foundation, Wistar Launches its First National, College-level Biomedical Research Internship Program

Inaugural class learns how to conduct research in immersive summer program

If you take a walk along any one of Wistar’s corridors in the summer, chances are you will see students at the laboratory bench working intently on their research projects. This summer is no exception and includes a select group of college students arriving in Philadelphia for a 12-week Wistar summer intensive research experience.

In the new Research Experiences for Undergraduates (REU) Program students are fully immersed in biomedical science experiments under the guidance of a Wistar mentor-scientist leading one of 33+ active labs at the Institute. Students conduct innovative research, applying state-of-the-science techniques to plan and execute experiments and advance their projects. REU is funded by the National Science Foundation (NSF), organized to encourage STEM student innovators to pursue graduate education and eventual careers that will advance the life sciences into new directions.

Of the inaugural group of 16 students, 12 identified as female and five identified as male. 10 students were funded by the NSF REU grant and six were funded through a PAsmart grant. All NSF REU students were from underrepresented groups and participating from California, Delaware, Florida, Maryland, Massachusetts, New Jersey, Pennsylvania, and Texas.

“Every day Wistar invests in pioneering research accomplished by our groundbreaking scientists. That investment includes training our next-gen scientists. These students will become leading scientists in the future and will be our answer and our success. Their discoveries will become tomorrow’s life-saving therapies,” said Dr. Kristy Shuda McGuire.

Since joining Wistar, dean of Biomedical Studies Dr. Shuda McGuire’s singular focus has been to expand education and training programs to impact as many as possible underserved and underrepresented students from the surrounding greater Philadelphia community and now, with the support of the NSF from around the country.

Inaugural REU students Rickelle Wescott and Kiara Garcia Castro shared their experiences in the REU Program where days are filled learning biomedical research lab methods. Nights are for relaxing (in communal housing provided through the Program) and reading science articles, deep dives into more research, and nailing down protocols for the next day’s experiments. Eat, sleep and repeat.

Originally from Puerto Rico, Kiara always aspired to a science job. She’s a rising junior biology major at Temple University. She knew she wanted to do research but did not know where to start.

“Wistar is training friendly and accommodating to everyone. My mentor emphasized it was okay to make mistakes and not feel bad when you mess up because you are learning how to fix your mistakes. He emphasized all researchers have gone through this—you aren’t born knowing how to do research,” said Kiara.

Their diligence in the Program pays off as these next-gen scientists push past their fears about undertaking hugely complex science.

“What’s been the most challenging was to understand and absorb all this scientific information in a short amount of time and then to apply it. I had to understand the purpose of everything: the different solutions, different terms and experiments. That was the difficult part. You just try to be quick on your feet,” said Rickelle.

Rickelle, who is on the pre-med track at Hampton University, is undertaking a variety of research experiments far different from anything she’s done so far in her college science curriculum.

“At Wistar, you feel part of something that is making breakthroughs. You feel like you can be someone who can make a discovery too,” said Rickelle.

Genomic Origami: Wistar Scientist Dr. Kavitha Sarma Studies How the Shape of Our Genes Impacts Disease

A Q&A with Dr. Kavitha Sarma

Dr. Kavitha Sarma runs an independent lab focused on nucleic acid structures called R-loops that contain both DNA and RNA and assist in gene expression. Dr. Sarma, associate professor in Wistar’s Gene Expression and Regulation Program, recently published a paper in Molecular Cell about genomic structures — specifically, G-quadruplexes and R-loops.

R-loops are bubble-like structures that can form in our DNA, and they can affect how genes are expressed — whether genes are turned on or off. G-quadruplexes form on the single-strand DNA of R-loops and can stabilize R-loops. In her research, Dr. Sarma found that R-loops and G-quadruplexes can influence the binding of a protein called CTCF, which helps fold and organize our DNA. This folding process is important for gene expression. If the genome is folded correctly, that allows genes to be expressed the way they should be. But if the genome is folded incorrectly, it can cause faulty patterns of gene expression, which can potentially lead to disease and cancer. R-loops and G-quadruplexes can play a role in cancer and disease by recruiting CTCF in a way that promotes faulty gene expression.

IN YOUR PAPER, YOU FOUND THAT R-LOOPS AND G4S HAD AN INTERESTING RELATIONSHIP WITH A CERTAIN MOLECULE. COULD YOU EXPLAIN THAT FINDING?

In every cell nucleus in your body, you have something like two meters of DNA, if you were to unravel it completely into one long double helix. Just to make genetic information physically fit in your body, the genome has to be compacted, and that needs to happen in every single cell, too.

There are many proteins that function in genome folding. We found that R-loops and G4s can influence the binding of one of these proteins – CTCF, which has a very important role in how the genome is folded.

This folding process, which also serves as a kind of information organization process, is important for how cells develop and specialize in our body. For example, the way a neuron’s genome is folded and expressed will be different from the genome folding and gene expression of a pancreatic cell because the two cell types fulfill different purposes. Epigenetic regulation from factors like genome folding allows for a diversity of gene expression — which, in turn, allows for a diversity of cell types and functions.

So, if a genome is folded correctly in the nucleus and the right regions are next to each other, that has a positive effect, and genes are expressed the way they should be. But if CTCF folds the genome incorrectly — for example, if R-loops and G4s form and facilitate CTCF binding to regions where it isn’t supposed to bind — we might see incorrect patterns of gene expression and the kinds of dysregulation you’d find in cancer and disease.

WHAT ARE THE PATHOGENIC IMPLICATIONS OF CTCF RECRUITMENT?

This finding, that G4s affect CTCF, tells us that the genome misfolding in disease can be at least partially due to the formation of R-loop structures. In addition to developmental disorders, R-loop and G4 structures can play problematic roles in cancer because they’re what we call co-transcriptional. When transcription happens, these structures tend to accumulate — they tend to become stabilized. Hypertranscription that occurs in many cancers can contribute to genome misfolding through R-loop and G4 formation, which can further reinforce faulty gene expression patterns by essentially rewiring the genome.

WHAT DOES THIS REWIRING CYCLE TELL US ABOUT THE EPIGENETICS OF CANCER AND DISEASE?

I think that this research gives us a good roadmap for looking for therapeutics down the line, because a better understanding of epigenetic regulation gives us deeper insight into how disease states work at a very localized level.

We know that R-loops and G4s can alter CTCF binding and change genome folding. Going forward, we can identify pathogenic contacts that occur because of these genomic structures and try to correct them. This is how foundational research — understanding processes that weren’t understood before — can lead to advances in the science of human health.

Aaron R. Goldman, Ph.D.

Assistant Professor, Molecular and Cellular Oncogenesis Program, Ellen and Ronald Caplan Cancer Center

Goldman applies mass spectrometry (MS)-based approaches, primarily metabolomics and lipidomics, to examine metabolism in a wide-range of biomedical studies applied to cancer and other human diseases as well as fundamental mechanistic biology.

Goldman received his B.S. in Biological Sciences from Carnegie Mellon University. He earned a Ph.D. in Cell and Molecular Biology from Stanford University. Goldman joined The Wistar Institute in 2014 as a postdoctoral fellow in David Speicher’s laboratory where he utilized mass spectrometry-based proteomics, lipidomics, and metabolomics to study melanoma, ovarian cancer, and other cancers. He was subsequently appointed as Associate Managing Director of Metabolomics and Lipidomics in the Proteomics and Metabolomics Shared Resource. In 2022, Goldman was promoted to Assistant Professor in the Molecular and Cellular Oncogenesis Program of Wistar’s Ellen and Ronald Caplan Cancer Center.

View Publications

The Goldman Laboratory

agoldman@wistar.org

The Goldman Laboratory

The Goldman laboratory extensively utilizes state-of-the-art high resolution mass spectrometry to study metabolic and lipid changes in cancer and infectious diseases in collaboration with researchers at Wistar and many other institutions. Most projects are systems-level studies that assimilate data from multiple sources to better understand the mechanisms underlying diseases, to identify potential therapeutic targets, and to uncover putative diagnostic and prognostic biomarkers.

Metabolomics and lipidomics are the study of the set of polar metabolites and lipids (non-polar metabolites), respectively, in a biological system. Unbiased profiling of these small molecules provides insight into metabolic pathways and processes perturbed in disease states that could lead to testable hypotheses for disease progression or therapy response. Alternatively, these methods can confirm findings suggested by orthogonal assays such as transcriptomics or proteomics. Use of these techniques has exponentially increased in recent years as mass spectrometry technologies have enabled greater depth of analysis and more confident identification of specific metabolites and lipids.

Available Positions

Motivated candidates are encouraged to inquire about the positions below by contacting Dr. Goldman at agoldman@wistar.org.

A Postdoctoral Fellow and a Research Assistant position are available to work on multiple ongoing projects in the Goldman laboratory. The research focus of these positions could include (depending upon interest and expertise): small molecule omics studies of melanoma tumor progression and resistance to therapies; pregnancy biomarker validation; and/or implementation of new and improved mass spectrometry-based analyses of polar metabolites and lipids (see project descriptions above). Postdoctoral Fellow candidates should have recently received or be close to obtaining their Ph.D. degree (or equivalent) and have a strong background in small molecule mass spectrometry. Research Assistant candidates should have a B.S. degree in biochemistry or equivalent. Experience with MS is preferred but not required.

Research

The Goldman laboratory is focused on improving both mass spectrometry-based metabolomics and lipidomics methods and applying these state-of-the-art methods to a wide-range of collaborative studies investigating cancers including ovarian, pancreatic, prostate, melanoma, and leukemia, as well as infectious diseases including HIV and COVID-19. A combination of untargeted and targeted approaches both at steady-state and using isotopic labeling (flux analysis) have led to important findings associated with disease progression and treatment, including changes in energy metabolism, perturbation of specific macromolecule biosynthesis pathways, and extensive lipid remodeling. Results of mass spectrometry studies have often been used to validate findings from RNAseq and other orthogonal approaches, and integration with other data sets has yielded combinatorial biomarkers.

Available Positions

Motivated candidates are encouraged to inquire about the positions below by contacting Dr. Goldman at agoldman@wistar.org.

A Postdoctoral Fellow and a Research Assistant position are available to work on multiple ongoing projects in the Goldman laboratory. The research focus of these positions could include (depending upon interest and expertise): small molecule omics studies of melanoma tumor progression and resistance to therapies; pregnancy biomarker validation; and/or implementation of new and improved mass spectrometry-based analyses of polar metabolites and lipids (see project descriptions above). Postdoctoral Fellow candidates should have recently received or be close to obtaining their Ph.D. degree (or equivalent) and have a strong background in small molecule mass spectrometry. Research Assistant candidates should have a B.S. degree in biochemistry or equivalent. Experience with MS is preferred but not required.

Major areas of interest:

LIPID REMODELING IN TUMOR PROGRESSION AND THERAPY RESISTANCE IN MELANOMA

A primary focus of the Goldman laboratory is collaborating with other researchers to define the role that lipid metabolism plays in melanoma progression and therapy resistance. In one such academic collaboration with researchers at the University of Pennsylvania, specific lipid metabolism pathways were implicated as being affected by lysosomal autophagy inhibition by using a combination of proteomic and lipidomic approaches. Efforts are ongoing to define the mechanisms by which autophagy modulation regulates lipid metabolism using small molecule activators and inhibitors. Combinatorial inhibition of lipid metabolism pathways and lysosomal autophagy is being pursued as a putative therapeutic strategy to mitigate therapy resistance in melanoma.

THE ROLE OF THE GUT MICROBIOME IN DISEASE

There is a growing field of research on the interplay between gut microbiota and human health. Recent work has found that the gut microbiome plays roles in tumor biology, infectious diseases, and immune response. Collaborative projects at Wistar led by the laboratories of Drs. Rahul Shinde and Mohamed Abdel-Mohsen have uncovered links between gut microbiota and gut-derived products with pancreatic cancer and COVID-19, respectively. Examples of key gut-derived products uncovered by untargeted metabolomic profiling are trimethylamine N-oxide, which is produced primarily from dietary choline, and various metabolites of tryptophan. The Goldman laboratory is actively developing and implementing methods to expand coverage of gut-derived products to 1) short-chain fatty acids (SCFA; two to six carbons) that regulate cell homeostasis and have been implicated in multiple cancers and other diseases such as HIV and 2) bile acids that are involved in lipid digestion and absorption and are subject to on-going research for their roles in cancer development and progression as well as potential therapeutic applications.

DISCOVERY AND VALIDATION OF EARLY PREGNANCY BIOMARKERS

A major clinical challenge in early pregnancy is to distinguish normal intrauterine pregnancy (IUP) from abnormal conditions when ultrasound is not diagnostic. Women with early-stage pregnancy who experience abdominal pain and vaginal bleeding might have: 1) an ongoing viable IUP, 2) a non-viable intrauterine pregnancy or spontaneous abortion (SAB), or 3) ectopic pregnancy (EP). EP occurs in 1-2% of pregnant women and causes 6% of pregnancy-related deaths, while SAB affects 10-20% of pregnancies. Because clinical treatment for these possible outcomes differs greatly, it is critical that an accurate diagnosis is made as early as possible. With researchers from the University of Pennsylvania, we are performing mass spectrometry-based studies to identify putative early pregnancy protein, metabolite and/or lipid biomarkers. The end goal is to develop a novel, multiplexed, MS-based plasma biomarker test for early and accurate diagnosis of pregnancy outcome. Targeted proteomics is being used to validate previously identified candidate biomarkers with a focus on distinguishing protein isoforms to increase accuracy, and discovery metabolomics and lipidomics are being pursued to identify additional targets that may complement protein markers in biomarker panels for early pregnancy outcomes.

EMERGING MS TECHNOLOGIES FOR SMALL MOLECULE STUDIES

Ion Mobility (IM). Goldman and his team are actively evaluating different IM implementations for metabolite and lipid studies. A major challenge in the analysis of small molecules is confident annotation of isomers (molecules with the same chemical formula) and isobars (molecules with the same nominal mass at a given resolution). These compounds are indistinguishable by accurate mass, and MS/MS fragmentation is often information-poor and does not generate diagnostic fragments that are unique to a given species. They may also not be resolved by chromatographic separation. A promising emerging technology to distinguish these types of compounds is IM, which separates ions in the gas phase based on their collisional cross-section (CCS), a property derived from the mobility of the ions in an electric field. In addition to separating currently unresolved species, IM provides a fourth dimension of data to annotate detected compounds more confidently in terms of retention time, accurate mass, MS/MS spectra, and CCS. This technology is also applicable to proteomics, where it can be used to separate isomeric peptides ¬– such as those that are phosphorylated on different residues – and reduce sample complexity to increase depth of analysis.

MS Imaging. Goldman and his team are evaluating alternative instruments and approaches for tissue imaging of metabolites, lipids, and other biomolecules. Tumors are heterogenous and determination of the spatial distribution of biomolecules may provide insights into disease states and allow for stratification of patients by tumor molecular signatures for tailored treatment options. Imaging mass spectrometry (MS) uses a matrix-assisted laser desorption/ionization (MALDI) source to visualize lipids, metabolites, and peptides directly or visualize proteins and glycans with additional processing (enzymatic digestion) at 5-10 µm resolution. Analyte patterns can be compared between conditions to identify important biomolecules and define molecular signatures. These data can be used to select regions for more in-depth study using laser microdissection followed by traditional LC-MS approaches and correlated with orthogonal assays such as immunohistochemistry. Recent instruments support IM and MS/MS fragmentation to allow for higher confidence annotation of analytes.

Selected Publications

  • A Cancer Ubiquitome Landscape Identifies Metabolic Reprogramming as Target of Parkin Tumor Suppression.

    Agarwal, E., Goldman, A.R., Tang, H.Y., Kossenkov, A.V., Ghosh, J.C., Languino, L.R., Vaira, V., Speicher, D.W., Altieri, D.C. “A Cancer Ubiquitome Landscape Identifies Metabolic Reprogramming as Target of Parkin Tumor Suppression.” Sci Adv. 2021 Aug 25;7(35):eabg7287. doi: 10.1126/sciadv.abg7287. Print 2021 Aug.

  • ATF3 Coordinates Serine and Nucleotide Metabolism to Drive Cell Cycle Progression in Acute Myeloid Leukemia.

    Di Marcantonio, D., Martinez, E., Kanefsky, J.S., Huhn, J.M., Gabbasov, R., Gupta, A., Krais, J.J., Peri, S., Tan, Y., Skorski, T., et al. “ATF3 Coordinates Serine and Nucleotide Metabolism to Drive Cell Cycle Progression in Acute Myeloid Leukemia.” Mol Cell. 2021 Jul 1;81(13):2752-2764.e6.doi: 10.1016/j.molcel.2021.05.008. Epub 2021 Jun 2.

  • Plasma Markers of Disrupted Gut Permeability in Severe COVID-19 Patients.

    Giron, L.B., Dweep, H., Yin, X., Wang, H., Damra, M., Goldman AR, Gorman N, Palmer CS, Tang HY, Shaikh MW, et al. “Plasma Markers of Disrupted Gut Permeability in Severe COVID-19 Patients.” Front Immunol. 2021 Jun 9;12:686240. doi: 10.3389/fimmu.2021.686240. eCollection 2021.

  • Changes in Aged Fibroblast Lipid Metabolism Induce Age-Dependent Melanoma Cell Resistance to Targeted Therapy via the Fatty Acid Transporter FATP2.

    Alicea, G.M., Rebecca, V.W., Goldman, A.R., Fane, M.E., Douglass, S.M., Behera, R., Webster, M.R., Kugel, C.H. 3rd, Ecker, B.L., Caino, M.C., et al. “Changes in Aged Fibroblast Lipid Metabolism Induce Age-Dependent Melanoma Cell Resistance to Targeted Therapy via the Fatty Acid Transporter FATP2.” Cancer Discov. 2020 Sep;10(9):1282-1295. doi: 10.1158/2159-8290.CD-20-0329. Epub 2020 Jun 4.

  • PPT1 Promotes Tumor Growth and Is the Molecular Target of Chloroquine Derivatives in Cancer.

    Rebecca, V.W., Nicastri, M.C., Fennelly, C., Chude, C.I., Barber-Rotenberg, J.S., Ronghe, A., McAfee, Q., McLaughlin, N.P., Zhang, G., Goldman, A.R., et al. “PPT1 Promotes Tumor Growth and Is the Molecular Target of Chloroquine Derivatives in Cancer.” Cancer Discov. 2019 Feb;9(2):220-229. doi: 10.1158/2159-8290.CD-18-0706. Epub 2018 Nov 15.

View Additional Publications

Continue reading

Alessandro Gardini, Ph.D.

Associate Professor, Genome Regulation and Cell Signaling Program, Ellen and Ronald Caplan Cancer Center

Gardini studies the epigenetic control of transcription during cell differentiation and oncogenesis.

Born and raised in Italy, Gardini obtained a B.S./M.S. in medical biotechnology at the University of Bologna and attended the graduate school of Molecular Medicine at the University of Milan. He trained as a postdoctoral fellow with Dr. Ramin Shiekhattar at the Center of Genomic Regulation in Barcelona, The Wistar Institute and the University of Miami Medical School. He joined Wistar as an assistant professor in 2015. Gardini is a scholar of the Leukemia Research Foundation and the American Cancer Society.

View Publications

The Gardini Laboratory

215-898-3785

agardini@wistar.org

The Gardini Laboratory

Transcriptional regulation is a multi-layer process carried out by specialized transcription factors, co-factors and enzymatic machineries working within the context of a structured chromatin landscape.

We use a combination of genomics, proteomics and biochemistry to investigate how basal transcriptional complexes and epigenetic modulators orchestrate gene expression in normal and tumor cells.

Staff
  • Postdoctoral Fellows

    Sandra Deliard, Ph.D.

  • Graduate Students

    Luca Grillini (UniBO)
    Connor Hill (UPenn-CAMB)
    Ilan Kirkel (UniBO)
    Sarah Offley (UPenn-CAMB)
    Martina Gatto (UniBO)

  • Research Assistant

    Francis Picone


Alumni

Marco Trizzino, Ph.D. (2016-2019), assistant professor, Thomas Jefferson University

Elisa Barbieri, Ph.D. (2015-2019), Marie Sklodowska-Curie Fellow, University of Edinburgh


Available Positions

Motivated candidates are encouraged to inquire about the positions below. Contact agardini@wistar.org.

Postdoctoral Fellow
Graduate Students (BGS-UPenn)

Research

  • 1 – Enhancer Regulation During Lineage and Tissue Specification

    We dissect the transcriptional mechanisms that control fate choice and differentiation of human hematopoietic cells, particularly in the myeloid compartment. We are especially interested in the role of transcriptional enhancers.

    Enhancers are distal regulatory elements, scattered throughout the entire genome, that direct gene regulation during development. Furthermore, enhancers are active spots for transcription of long noncoding eRNAs (Gardini and Shiekhattar, 2015). These short-lived, non-polyadenylated transcripts are required to establish a link between the promoter and the distant regulatory enhancer (chromosomal looping) and contribute to the expression of the target protein-coding gene. The underlying mechanisms are poorly understood and the potential contribution of enhancers and eRNAs to cancer has just started to emerge. The lab is keen on understanding how enhancer networks get activated and promote differentiation of myeloid cells, and how this process is disrupted during leukemogenesis. We recently uncovered a novel enhancer regulatory axis in myeloid progenitor cells (Barbieri, Trizzino et al., 2018), centered around the EGR-1 transcription factor and a newly characterized module of the Integrator complex.

    Additional studies (Trizzino, Zucco et al., 2021) revealed that EGR-1 also coordinates a repressive activity, independent of Integrator, in mature macrophages. Particularly, EGR-1 restrains the activity of inflammatory enhancers and effectively curbs the response of macrophages to inflammatory stimuli.

    Download the image here.

  • 2 – Role of Protein Phosphatases in RNA Polymerase Pause-Release and Elongation

    Eukaryotic transcription pivots around the balanced activity of kinases and phosphatases that regulate the transcription initiation, early elongation and termination checkpoints. While the dynamic recruitment of kinases (i.e. CDK7, CDK9) and their cognate cyclins have been studied for many years, the association of phosphatases with active transcription has been largely understudied. We recently uncovered that protein phosphatase 2A (PP2A) is recruited at the pause-release checkpoint where it opposes CDK9 activity (Vervoort, Welsh et al., 2021). We found that the Integrator subunit INTS6 draws PP2A to the transcription bubble to dephosphorylate the RNAPII-CTD (especially Ser2) and maintain polymerase in a paused conformation. PP2A activity is frequently lost in cancer and restoring its ability to curb transcription opens up new therapeutic strategies for tumors ‘addicted’ to transcriptional elongation.

    Download the image here.

  • 3 – Integrator: The Swiss Army Knife of Transcription

    Integrator is a large, evolutionarily conserved, multiprotein complex that is quickly emerging as a centerpiece of transcriptional regulation in higher eukaryotes (Welsh and Gardini, 2022). The Integrator complex is a regulatory hub for several transcriptional and epigenetic processes. For instance, Integrator’s INTS11 subunit controls release of RNA Polymerase II at promoters via endonucleolytic cut of nascent RNA (Gardini et al., 2014Beckedorff et al., 2020). In addition, INTS11 regulates enhancer function through the processing of noncoding eRNAs (Lai, Gardini et al., 2015). We identified a new functional module of Integrator, centered around the INTS13 subunit, that prompts enhancer activation in myeloid cells (Barbieri, Trizzino et al., 2018). More recently, we showed that the INTS6/INTS8 subunits of the complex recruit the PP2A phosphatase to chromatin, to control phosphorylation of the RNAPII-CTD as well as other polymerase co-factors implicated in pausing and elongation (Vervoort, Welsh et al., 2021). Notably, Integrator subunits are found mutated in developmental diseases and are frequently lost in certain tumor types. The lab keeps pursuing the biochemical and functional dissection of all 14 subunits of the Integrator complex.

    Download the image here.

  • 4 – Profiling Human Transcriptomes at Better Resolution

    Conventional transcriptomic analyses (i.e. ribo-depleted RNA-seq, 3’-seq) offer a glimpse into global gene regulation but often fail to convey the most accurate picture. Because RNA-seq captures steady-state and cytoplasmic transcripts, changes in gene expression can be hindered by RNA stability. Furthermore, RNA Polymerase dynamics is only revealed by real-time nascent RNA transcription. We strive to implement and optimize techniques of nascent RNA-seq, improving their reproducibility and applicability. To this purpose, we recently developed fastGRO (Barbieri, Hill et al., 2020) as a global nuclear run-on assay of rapid execution and suitable for primary cells.

    Download the image here.

  • 5 – Role of ARID1A in Transcriptional Regulation

    Chromatin remodelers regulate DNA accessibility by positioning nucleosomes at active promoters and enhancers. We investigate the function of SWI/SNF, the most mutated chromatin remodeler across all human cancers. Subunits of SWI/SNF, such as ARID1A, are found mutated in sporadic ovarian tumors and their specific contribution to SWI/SNF activity is poorly understood. We recently elucidated a novel function of ARID1A in pausing of RNA Polymerase II (Trizzino et al., 2018).

Gardini Lab in the News

Selected Publications

  • The PP2A-Integrator-CDK9 Axis Fine-tunes Transcription and Can Be Targeted Therapeutically in Cancer.

    Vervoort, S.J., Welsh, S.A., Devlin, J.R., Barbieri, E., Knight, D.A., Offley, S., Bjelosevic, S., Costacurta, M., Todorovski, I., Kearney, C.J., et al. “The PP2A-Integrator-CDK9 Axis Fine-tunes Transcription and Can Be Targeted Therapeutically in Cancer.” Cell. 2021 May 17; doi: 10.1016/j.cell.2021.04.022

  • EGR1 is a Gatekeeper of Inflammatory Enhancers in Human Macrophages

    Trizzino, M., Zucco, A., Deliard, S., Wang, F., Barbieri. E., Veglia, F., Gabrilovich, D., Gardini, A. “EGR1 is a Gatekeeper of Inflammatory Enhancers in Human Macrophages” Sci Adv. 2021 Jan 13;7(3):eaaz8836. doi: 10.1126/sciadv.aaz8836. Print 2021 Jan.

  • Rapid and Scalable Profiling of Nascent RNA with fastGRO

    Barbieri, E., Hill, C., Quesnel-Vallières, M., Zucco, A.J., Barash, Y., Gardini, A. “Rapid and Scalable Profiling of Nascent RNA with fastGRO” Cell Rep. 2020 Nov 10;33(6):108373. doi:10.1016/j.celrep.2020.108373.

  • Targeted enhancer activation by a subunit of the Integrator complex

    Barbieri, E., Trizzino, M., Welsh, S.A., Owens, T.A., Calabretta, B., Carroll, M., Sarma, K., Gardini, A. “Targeted enhancer activation by a subunit of the Integrator complex.” Mol Cell. 2018. 

  • The tumor suppressor ARID1A controls global transcription via pausing of RNA Polymerase II.

    Trizzino, M., Barbieri, E., Petracovici, A., Wu, S., Welsh, S.A., Owens, T.A., Licciulli, S., Zhang, R., Gardini, A. “The tumor suppressor ARID1A controls global transcription via pausing of RNA Polymerase II.” Cell Reports. 2018.

View Additional Publications

Continue reading

Amelia Escolano, Ph.D.

Assistant Professor, Vaccine & Immunotherapy Center

The Escolano Lab investigates novel vaccination strategies against highly mutating viruses.

We are interested in understanding the unique features of the humoral and cellular immune responses to sequential immunization, with special focus on the process of antibody affinity maturation in the germinal centers. Our goal is to rationally design vaccination approaches to induce potent and long-lasting antibody responses against pathogens that diversify over time.

Dr. Amelia Escolano is an Assistant Professor in the Vaccine & Immunotherapy Center and a Wistar Institute Assistant Professor in Microbiology at the University of Pennsylvania.

Escolano obtained her BS degree in Biochemistry from the University of Oviedo, Spain and a master’s degree from Centro de Biologia Molecular Severo Ochoa in Madrid, Spain. She received additional training at the University of Turku, Finland and the Genome Research Institute in Cincinnati, Ohio. Escolano obtained her PhD in biochemistry and molecular biology from Autonoma University of Madrid after completing her pre doctoral studies at the Spanish National Center for Cardiovascular Research (CNIC) in Madrid. She trained as a postdoctoral fellow in the laboratory of Michel Nussenzweig at The Rockefeller University in New York and joined The Wistar Institute as an Assistant Professor in 2021. Escolano is a Pew Biomedical Scholar, a recipient of the regional Blavatnik award for young scientists (finalist) and has been recognized with a NIH Director’s New Innovator Award (DP2).

View Publications

The Escolano Laboratory

215-898-3703

aescolano@wistar.org

The Escolano Laboratory

Viruses such as HIV-1, influenza and SARS-CoV-2 have the ability to rapidly mutate as a mechanism to escape from the host immune system. As a result, highly mutating viruses are largely diverse, with multiple different circulating variants characterized by individual antigenic and infectivity properties. The design of efficacious vaccines against highly diverse viruses is extraordinarily challenging. Despite decades of research, no universal vaccines exist against HIV-1 or influenza. Unfortunately, common vaccination strategies cannot elicit the type of broadly neutralizing antibodies required to confer broad protection against these viruses, and some of the currently available vaccines fail to induce long-lasting protection. Consequently, boost immunizations are advised in order to achieve protection.

The Escolano Lab investigates the use of sequential immunization as a novel vaccination strategy aiming to induce broadly neutralizing antibodies against highly diverse viruses. Using HIV-1 as a model virus, state-of-the-art technologies and animal models including wild type mice, transgenic mice and rhesus macaques, the laboratory studies the humoral and cellular immune responses to sequential immunization. The research goal is to identify guidelines for the design of vaccination approaches to induce long-lasting protection against highly mutating viruses in humans.

Staff
  • Postdoctoral Fellows

    Ignacio Rodriguez Relaño, Ph.D.
    Maria Belen Palacio, Ph.D.
    Marta Tarquis, Ph.D.

  • Ph.D. Student (UniBo)

    Zainul Abe Din

  • Graduate Students

    Ashwin Skelly
    Austin Kriews

  • Research Assistant

    Caroline Boroughs
    Sowmya Meka
    Maggie Kerwin


Available Positions

Postdoctoral fellow and research assistant positions are available in the Escolano laboratory. Interested applicants are encouraged to contact aescolano@wistar.org.

Research

  • Sequential Immunization Against HIV-1

    An efficacious antibody-based vaccine against HIV-1 should elicit broadly neutralizing antibodies (bNAbs) targeting conserved epitopes of its envelope (Env) protein. Our previous work showed that common vaccination strategies using repeated boost immunization with the same Env immunogen could not elicit bNAbs. Instead, a new form of vaccination involving sequential immunization was required to elicit highly mutated anti-HIV-1 bNAbs. The reported sequential immunization protocol involved prime immunization with an engineered Env immunogen followed by a series of four additional immunizations with Env immunogens that gradually resembled the native-looking Env. This immunization protocol elicited bNAbs in an immunoglobulin knock-in mouse model with a monoclonal B cell repertoire engineered to carry the inferred germline precursor of a human bNAb (Escolano et al, Cell, 2016). These immunization experiments were the first showing that anti-HIV-1 bNAbs can be elicited by vaccination, and fueled subsequent vaccine design studies. However, despite this significant achievement, no vaccination protocols have been reported that are able to elicit protective levels of bNAbs in wild type organisms with a polyclonal B cell repertoire.

    Wild type organisms mount polyclonal antibody responses when encountering complex antigens such as the HIV-1 Env protein. A predominant component of antibody responses elicited by Env immunogens are antibodies to non-conserved or strain-specific epitopes of Env, with no potential to broadly neutralize HIV-1. These antibodies significantly interfere with the development of bNAbs.

    The Escolano lab investigates the humoral and cellular immune responses to sequential immunization to establish guidelines for vaccine design. We design and evaluate immunogens and sequential immunization strategies to elicit anti HIV-1 broadly neutralizing antibodies in wild type organisms. Our specific goal is to devise approaches to modulate the immunodominance properties of vaccine candidates aiming to focus the antibody responses to the conserved, neutralization-sensitive epitopes of Env.

    Using state-of-the-art technologies for single cell analysis and different animal models including wild type mice, rhesus macaques and a series of recently generated immunoglobulin knock-in and reporter mice, we investigate the process of antibody maturation and the evolution of the different immune compartments upon sequential immunization.

  • Antibody Isolation and Characterization

    Our laboratory has vast experience designing strategies to isolate antigen-specific B cells from wild type mice, humanized mice and non-human primates, and cloning their antibody genes.

    Antibody cloning from single B cells is an essential tool for characterizing humoral immune responses and obtaining valuable therapeutic and analytical reagents. Antibody cloning from individuals with high serologic titers to HIV-1, influenza, malaria, ZIKV, and SARS-CoV-2 has led to new insights that inform vaccine design efforts.

    We have designed cost-effective protocols to identify and purify single antigen-specific B cells, and subsequently clone and produce monoclonal antibodies.

    We are using the newly developed methods to isolate and characterize HIV-1-specific B cells from naïve and immunized mice, from vaccinated or simian-human immunodeficiency virus (SHIV)-infected rhesus macaques and from humans. Remarkably, using our approach, we have isolated one of the first anti-HIV-1 bNAbs from a SHIV-infected rhesus macaque, validating the use of macaques as preclinical models for HIV-1 vaccination studies.

    The Escolano laboratory uses this state-of-the-art methodology to isolate antibodies against bacteria, tumor neoantigens and viruses, including cancer-associated viruses. Characterization of the isolated antibodies is providing very valuable information to guide vaccine design efforts and the development of new preventative and therapeutic approaches.

  • Production Of Knock-in Mice

    The use of animal models in biomedical research has been crucial to investigate the mechanisms of human pathophysiology, evaluate candidate interventions and predict treatment outcomes in humans. Animal Models have been extensively used by the scientific community to examine the cellular and humoral responses to infection and vaccination. Unfortunately, none of these models faithfully recapitulates the setting of a human immune response.

    Humanized mice genetically engineered to recapitulate different aspects of the human humoral and/or cellular immune response are highly desirable. In particular, human immunoglobulin knock-in mice (Ig KI mice) are remarkably valuable for vaccine development and drug discovery, as well as for basic immunology studies related to the analysis of B and T cell responses to infection, vaccination, autoimmunity, or cancer. However, current methods to produce Ig KI mice are inefficient, labor-intensive and require special equipment and expertise to perform zygote microinjections.

    We have developed a novel technology to efficiently and more easily generate monoclonal Ig KI mice using CRISPR/Cas9. The new technology remarkably simplifies the mouse production process, increases knock-in efficiency and reduces breeding time, thus accelerating the production process and reducing cost.

    We are currently using the new technology for high-throughput production of Ig KI mice carrying anti-HIV-1 antibodies, which we use for vaccine design purposes.

    Interestingly, this methodology can be adapted to introduce other genetic modifications in the mouse genome including gene insertions and deletions, thus being of great value to other scientific disciplines. The new technology is especially valuable for engineering events involving insertions of long DNA fragments or genetic modification of more than one locus.

Staff
  • Postdoctoral Fellows

    Ignacio Rodriguez Relaño, Ph.D.
    Maria Belen Palacio, Ph.D.
    Marta Tarquis, Ph.D.

  • Ph.D. Student (UniBo)

    Zainul Abe Din

  • Graduate Students

    Ashwin Skelly
    Austin Kriews

  • Research Assistant

    Caroline Boroughs
    Sowmya Meka
    Maggie Kerwin


Available Positions

Postdoctoral fellow and research assistant positions are available in the Escolano laboratory. Interested applicants are encouraged to contact aescolano@wistar.org.

Escolano Lab in the News

  • From Forbes: Meet The 2022 Class Of Pew Scholars In Biomedical Sciences

  • Wistar’s Dr. Amelia Escolano Earns NIH Director’s New Innovator Award

  • Wistar’s Dr. Amelia Escolano Named 2022 Pew Scholar

Selected Publications

  • Sequential Immunization Elicits Broadly Neutralizing Anti-HIV-1 Antibodies in Ig Knockin Mice.

    Escolano, A., Steichen, J.M., Dosenovic, P., Kulp, D.W., Golijanin, J., Sok, D., Freund, N.T., Gitlin, A.D. Oliveira, T. Araki, T., et al. “Sequential Immunization Elicits Broadly Neutralizing Anti-HIV-1 Antibodies in Ig Knockin Mice.” Cell. 2016 Sep 8;166(6):1445-1458.e12. doi: 10.1016/j.cell.2016.07.030.

  • Immunization Expands B Cells Specific To HIV-1 V3 Glycan In Mice And Macaques.

    Escolano, A., Gristick, H.B., Abernathy, M.E., Merkenschlager, J., Gautam, R., Oliveira, T.Y., Pai, J., West Jr, A.P., Barnes, C.O., Cohen, A.A., et al. “Immunization Expands B Cells Specific To HIV-1 V3 Glycan In Mice And Macaques.” Nature. 2019 Jun;570(7762):468-473. doi: 10.1038/s41586-019-1250-z. Epub 2019 May 29.

  • Sequential immunization of macaques elicits heterologous, neutralizing antibodies targeting the V3-glycan patch of HIV-1 Env.

    Escolano A, Gristick HB, Gautam R, DeLaitsch AT, Abernathy ME, Yang Z, Wang H, Hoffmann MAG, Nishimura Y, Wang Z, Koranda N, Kakutani LM, Gao H, Gnanapragasam PNP, Raina H, Gazumyan A, Cipolla M, Oliveira TY, Ramos V, Irvine DJ, Silva M, West AP Jr, Keeffe JR, Barnes CO, Seaman MS, Nussenzweig MC, Martin MA, Bjorkman PJ. Sequential immunization of macaques elicits heterologous neutralizing antibodies targeting the V3-glycan patch of HIV-1 Env. Sci Transl Med. 2021 Nov 24;13(621):eabk1533. doi: 10.1126/scitranslmed.abk1533. Epub 2021 Nov 24. PMID: 34818054; PMCIDPMC8932345.

  • A Broadly Neutralizing Macaque Monoclonal Antibody Against The HIV-1 V3-Glycan Patch.

    Wang, Z., Barnes, C.O., Gautam, R., Cetrulo Lorenzi, J.C., Mayer, C.T., Oliveira, T.Y., Ramos, V., Cipolla, M., Gordon, K.M., Gristick, H.B., et al. “A Broadly Neutralizing Macaque Monoclonal Antibody Against The HIV-1 V3-Glycan Patch.” Elife. 2020 Oct 21;9:e61991. doi: 10.7554/eLife.61991.

  • Progress Toward Active Or Passive HIV-1 Vaccination.

    Escolano, A., Dosenovic, P., Nussenzweig , M.C. “Progress Toward Active Or Passive HIV-1 Vaccination.” J Exp Med. 2017 Jan;214(1):3-16. doi: 10.1084/jem.20161765. Epub 2016 Dec 21.

View Additional Publications

Continue reading

Hildegund C.J. Ertl, M.D.

Professor, Vaccine & Immunotherapy Center

Ertl’s research centers on developing vaccines for an array of diseases and conditions—including AIDS, chronic viral infections, COVID-19 and some forms of cancer—not typically considered to be treated using this approach. These vaccines aim to protect against future infections and look to create new therapies for diseases already affecting people.

Ertl came to The Wistar Institute as an associate professor in 1987. A native of Germany, she received her medical degree from University of Göttingen. While in medical school, she began her scientific training as a student in the Max Planck Institute of Experimental Medicine. After research fellowships at the Australian National University and the University of Minnesota, Ertl joined the faculty of Harvard University before transitioning to Wistar. She became a full professor at Wistar in 1996 and holds professorships at the University of Pennsylvania School of Medicine and The Children’s Hospital of Philadelphia.

View Publications

The Ertl Laboratory

215-898-3863

ertl@wistar.org

The Ertl Laboratory

The Ertl laboratory has pioneered numerous patented technologies to create new vaccines. Much of the laboratory’s efforts focus on developing a new preventative vaccine for rabies—a lethal and underestimated disease that remains a top public health priority across the globe—have yielded useful technologies that the Ertl laboratory is applying to combating other viruses.

The lab is applying its adenovirus vaccine expertise against SARS-CoV-2, utilizing a modified chimpanzee virus as a vaccine delivery vehicle to induce an immune response. They are applying this platform to develop HIV vaccines and creating new therapeutic vaccines against human papillomavirus (HPV), a leading cause of cervical cancer, and chronic hepatitis B virus infection, a major cause of hepatocellular carcinoma.

Staff
  • Senior Staff Scientists

    Zhi Quan Jason Xiang, M.D.
    Xiang Yang Peter Zhou, M.D., Ph.D.

  • Postdoctoral Fellows

    Mohadeseh Hasanpourghadi, Ph.D.
    Mohsen Mohammadi, Ph.D.

  • Wistar Research Assistants

    Wynetta Giles-Davis
    Amara Saha

  • Lab Coordinator

    Christina Cole

Research

Vaccines to SARS-COV-2

We developed vaccines expressing the SARS-CoV-2 spike or nucleocapsid protein. The vaccines are being tested for T and B cell responses in mice. They are being tested in a hamster challenge model.

Therapeutic Cancer Vaccine to Human Papilloma Virus

Vaccines that aim to expand tumor-specific CD8(+) T cells have yielded disappointing results in cancer patients although they showed efficacy in transplantable tumor mouse models. Using a system that more faithfully mimics a progressing cancer and its immunoinhibitory microenvironment, we show that in transgenic mice, which gradually develop adenocarcinomas due to expression of HPV-16 E7 within their thyroid, a highly immunogenic vaccine expressing E7 only induces low E7-specific CD8(+) T-cell responses, which fail to affect the size of the tumors.

In contrast, the same type of vaccine expressing E7 fused to herpes simplex virus (HSV)-1 glycoprotein D (gD), an antagonist of the coinhibitory B- and T-lymphocyte attenuator (BTLA)/CD160-herpes virus entry mediator (HVEM) pathways, stimulates potent E7-specific CD8(+) T-cell responses, which can be augmented by repeated vaccination, resulting in initial regression of even large tumor masses in all mice with sustained regression in more than half of them. These results indicate that active immunization concomitantly with blockade of the immunoinhibitory HVEM-BTLA/CD160 pathways through HSV-1 gD may result in sustained tumor regression.

HIV-1 Vaccine Based on Chimp Serotypes of Adenovirus

This NIH-funded research aimed to test adenoviral recombinants based on simian serotypes for induction of immune responses to gag/pol/rev/env of HIV-1 or SIV-1 in mouse and primate models. Results were promising and part of the vaccines, based on two adenoviral vector vaccines expressing the HIV-1 env protein, are currently being tested in a phase I trial sponsored by HVTN.

Genetic Vaccine to Rabies Virus

The laboratory developed an adenovirus-based vaccine against rabies virus that can provide rapid immunity following a single administration. A simian adenoviral vector termed adenovirus C68 (AdC68) was generated as a molecular clone to express the glycoprotein of rabies virus. In mice, this vector induced complete protection to rabies virus challenge after a single dose. This vaccine also achieves long-term protection in non-human primates after a single dose. A slightly modified version of this vaccine has been tested in collaboration with The University of Oxford. Results were promising and a phase 1b/II trial is being initiated.

Therapeutic HBV Vaccine Based on Chimp Serotypes of Adenovirus

In collaboration with Virion Therapeutics, the laboratory developed a therapeutic vaccine against chronic hepatitis B virus (HBV) infection (CHB). The vaccines target viral polymerase and core proteins. Vaccines are delivered by chimpanzee adenovirus vectors (AdC) of serotype 6 (AdC6) and 7 (AdC7) used in prime-only or prime-boost regimens. The HBV antigens are fused into an early T cell checkpoint inhibitor, i.e., herpes simplex virus (HSV) glycoprotein D (gD), which enhances and broadens vaccine induced CD8+ T cell responses. The vaccines were shown to induce potent CD8+ T cells in mice which can reduce HBV viral loads in a surrogate model of CHB. Clinical trials are planned and expected to commence towards the end of 2022.

Metabolic Manipulation of CD8+ T Cells to Enhance Their Ability to Delay Tumor Progression

Reducing the metabolic stress within a tumor microenvironment could be essential to improve the effectiveness of active cancer immunotherapy. Using a mouse model of melanoma, the laboratory showed that appropriately timed treatment with the PPAR agonist fenofibrate improves the ability of a T cell-inducing cancer vaccine to delay tumor progression. The drug reduces the use of glucose by tumor and tumor stroma cells and promotes the use of fatty acids for their metabolic needs. The increased availability of glucose within the tumor microenvironment in turn allows for its increased use by vaccine-induced tumor-infiltrating CD8+ T cells, which improves their ability to slow tumor progression. The laboratory is currently using a humanized mouse model to test if similar results can be obtained with human melanomas.

Ertl Lab in the News

Selected Publications

  • Immunological Biomarker Discovery in Cure Regimens for Chronic Hepatitis B Virus Infection.

    Gehring, A.J., Mendez, P., Richter, K., Ertl, H., Donaldson, E.F., Mishra, P., Maini, M., Boonstra, A., Lauer, G., Creus, A., et al. “Immunological Biomarker Discovery in Cure Regimens for Chronic Hepatitis B Virus Infection.” J Hepatol. 2022 Mar 5;S0168-8278(22)00127-1. doi: 10.1016/j.jhep.2022.02.020.

  • The Effect of Rapamycin and Ibrutinib on Antibody Responses to Adeno-associated Virus Vector-mediated Gene Transfer.

    Xiang, Z., Kuranda, K., Quinn, W., Chekaoui, A., Ambrose, R., Hasanpourghadi, M., Novikov, M., Newman, D., Cole, C., Zhou, X., et al. “The Effect of Rapamycin and Ibrutinib on Antibody Responses to Adeno-associated Virus Vector-mediated Gene Transfer.” Hum Gene Ther. 2022 Mar 1. doi: 10.1089/hum.2021.258. Online ahead of print.

  • Hepatitis B Virus Polymerase-specific T cell Epitopes Shift in a Mouse Model of Chronic Infection.

    Hasanpourghadi, M., Novikov, M., Newman, D., Xiang, Z., Zhou, X.Y., Magowan, C., Ertl, H.C.J. “Hepatitis B Virus Polymerase-specific T cell Epitopes Shift in a Mouse Model of Chronic Infection.” Virol J. 2021 Dec 7;18(1):242. doi: 10.1186/s12985-021-01712-y.

  • PPARα Agonist Fenofibrate Enhances Cancer Vaccine Efficacy.

    Chekaoui, A., Ertl, H.C.J. “PPARα Agonist Fenofibrate Enhances Cancer Vaccine Efficacy.” Cancer Res. 2021 Sep 1;81(17):4431-4440. doi: 10.1158/0008-5472.CAN-21-0052. Epub 2021 Jul 8.

  • COVID-19 Vaccines Based on Adenovirus Vectors.

    Hasanpourghadi, M., Novikov, M., Ertl, H.C.J. “COVID-19 Vaccines Based on Adenovirus Vectors.” Trends Biochem Sci. 2021 May;46(5):429-430. doi: 10.1016/j.tibs.2021.03.002.

View Additional Publications

Continue reading

Daniel Claiborne, Ph.D.

  • Assistant Professor, Vaccine & Immunotherapy Center

  • Genome Regulation and Cell Signaling Program, Ellen and Ronald Caplan Cancer Center

  • Scientific Director, Histotechnology Facility

Claiborne is an immunologist focused on understanding how the function of T cells is modulated to create improved immunotherapies, including CAR T cell therapies, against HIV.

Claiborne earned his B.S. in Biochemistry from Florida State University and a Ph.D. in Immunology and Molecular Pathogenesis from Emory University. He completed his postdoctoral training at the Ragon Institute of MGH, MIT, and Harvard and joined The Wistar Institute in 2021 as a Caspar Wistar Fellow.

View Publications

The Claiborne Laboratory

215-898-2203

dclaiborne@wistar.org

The Claiborne Laboratory

T cell immunotherapies offer hope for the treatment of malignancies and other chronic diseases. However, natural mechanisms in place to limit immune-mediated pathology ultimately attenuate the T cell response in the setting of chronic antigen exposure, such as is found in chronic viral diseases – including HIV – as well as cancer. A better understanding of the mechanisms underlying T cell exhaustion has the potential to revolutionize T cell immunotherapies, such as chimeric antigen receptor (CAR) T cell therapy, for the treatment of human disease.

The Claiborne lab is rooted in understanding the complex interplay between the virus and the host in HIV transmission, pathogenesis, and persistence. A central focus of the lab is the optimization of CAR T cell therapy for a functional HIV cure. Concordant with efforts to engineer potent CAR T cell therapies against HIV, the Claiborne lab seeks to uncover the mechanisms behind the initiation and maintenance of dysfunctional T cell phenotypes, as the pursuit of these questions is critical to developing the next generation of T cell immunotherapies. To answer these complex questions, the lab leverages the powerful in vivo model system of HIV-infected humanized mice coupled with HIV-directed CAR T cells to study the evolution of antigen-specific T cell function over time.

Staff
  • Research Assistants

    Tyler Yang

  • Graduate Students

    Reyes Acosta (UPenn-CAMB)
    Alexandria Criswell (Drexel)
    Federica Severi (UniBO)

  • Postdoctoral Fellows

    Francesco Pennino, Ph.D.
    Nur Izzah Binti Ismail, Ph.D.

Research

MAPPING THE ONTOGENY OF DYSFUNCTIONAL CAR T CELL PHENOTYPES

Progressive T cell exhaustion and dysfunction due to continuous antigen exposure is a hallmark of chronic viral infections. In situations of uncontrolled HIV replication, we have observed a striking and progressive increase in the co-expression of multiple inhibitory receptors on CAR T cells in humanized mice. Additionally, CAR T cells isolated from viremic humanized mice display attenuated effector function ex vivo when compared to the transferred T cell product at baseline. Using this model system, and through a combination of transcriptomics and ex vivo functional assays, the Claiborne lab intends to map the initiation and maintenance of dysfunctional T cell subsets in an effort to uncover gene pathways/programs that can be targeted to prevent or reverse T cell exhaustion – with the goal of engineering more potent CAR T cell immunotherapies.

MAPPING THE KINETICS, CO-EXPRESSION, AND PLASTICITY OF INHIBITORY RECEPTOR EXPRESSION IN CHRONICALLY STIMULATED CAR T CELLS

We have recently developed a novel in vitro model system for chronic antigen stimulation of CAR T cells with the ability to specifically titrate antigen dosing as well as remove antigen stimulation at any time. Using this unique model system, we have mapped the longitudinal and concomitant expression of multiple inhibitory receptors on human CD4 and CD8 CAR T cells. We further aim to understand the functional consequences of multiple inhibitory receptor expression, whether periods of rest due to antigen removal can reverse certain inhibitory receptor profiles, and which profiles are indicative of long-term transcriptional reprogramming.

INFLUENCE OF VIRAL CHARACTERISTICS ON HIV-SPECIFIC T CELL FUNCTION

Previous work by the lab has demonstrated that the intrinsic viral replicative capacity (vRC) of the transmitted/founder virus can greatly impact the disease course of HIV-infected individuals. Individuals infected with high-vRC variants display exacerbated immunopathology characterized by T cell activation and exhaustion, concomitant with rapid CD4+ T cell loss. This may also represent an unrecognized hurdle for a functional HIV cure, as individuals with high-vRC viruses may be more refractory to CAR T cell therapy or other interventions. The lab endeavors to use a previously generated suite of chimeric viruses exhibiting distinct vRC phenotypes to define the extent to which vRC affects CAR T efficacy in vitro and in vivo and to elucidate the mechanisms responsible.

ROLE OF THE INNATE IMMUNE SYSTEM IN MODULATING CAR T CELL FUNCTION

Recent evidence in a humanized mouse model of acute lymphoblastic leukemia demonstrates that interactions between CAR T cells and the innate immune system can enhance CAR T cell function but can also contribute to cytokine release syndrome (Norelli M et al., Nature Medicine, 2018). During HIV infection, vigorous innate immune responses ultimately contribute to chronic immune activation and inflammation, which is associated with accelerated pathogenesis. These data suggest a complex role for components of the innate immune system, with the potential for enhancing or attenuating functions, in modulating CAR T cell efficacy in the context of HIV infection. We have created a series of reagents to specifically ablate or enhance monocyte function in humanized mice and endeavor to use this in vivo system to elucidate the role of the innate immune system in affecting HIV-specific CAR T cell function.

Claiborne Lab in the News

Selected Publications

  • Dual CD4-based CAR T Cells With Distinct Costimulatory Domains Mitigate HIV Pathogenesis In Vivo.

    Maldini, C.R., Claiborne, D.T., Okawa, K., Chen, T., Dopkin, D.L., Shan, X., Power, K.A, Trifonova, R.T., Krupp, K., Phelps, M., et al. “Dual CD4-based CAR T Cells With Distinct Costimulatory Domains Mitigate HIV Pathogenesis In Vivo.” Nat Med. 2020 Nov;26(11):1776-1787. doi: 10.1038/s41591-020-1039-5. Epub 2020 Aug 31.

  • Innate Immune Reconstitution in Humanized Bone Marrow-Liver-Thymus (HuBLT) Mice Governs Adaptive Cellular Immune Function and Responses to HIV-1 Infection.

    Garcia-Beltran, W.F., Claiborne, D.T., Maldini, C.R., Phelps, M., Vrbanac, V., Karpel, M.E., Krupp, K.L., Power, K.A., Boutwell, C.L., Balazs, A.B., et al. “Innate Immune Reconstitution in Humanized Bone Marrow-Liver-Thymus (HuBLT) Mice Governs Adaptive Cellular Immune Function and Responses to HIV-1 Infection.” Front Immunol. 2021 May 26;12:667393. doi: 10.3389/fimmu.2021.667393. eCollection 2021.

  • Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia.

    Claiborne, D.T. , Dudek, T.E., Maldini, C.R., Power, K.A., Ghebremichael, M., Seung, E., Mellors, E.F., Vrbanac, V.D., Krupp, K., Bisesi, A., et al. “Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia.” J Virol. 2019 Sep 30;93(20):e00814-19. doi: 10.1128/JVI.00814-19. Print 2019 Oct 15.

  • Replicative Fitness of Transmitted HIV-1 Drives Acute Immune Activation, Proviral Load in Memory CD4+ T Cells, and Disease Progression.

    Claiborne, D.T., Prince, J.L., Scully, E., Macharia, G., Micci, L., Lawson, B., Kopycinski, J., Deymier, M.J., Vanderford, T.H., Nganou-Makamdop, K., et al. ”Replicative Fitness of Transmitted HIV-1 Drives Acute Immune Activation, Proviral Load in Memory CD4+ T Cells, and Disease Progression.” Proc Natl Acad Sci U S A. 2015 Mar 24;112(12):E1480-9. doi: 10.1073/pnas.1421607112. Epub 2015 Feb 17.

  • Role Of Transmitted Gag CTL Polymorphisms In Defining Replicative Capacity And Early HIV-1 Pathogenesis.

    Prince, J.L., Claiborne, D.T., Carlson, J.M., Schaefer, M., Yu, T., Lahki, S., Prentice, H.A., Yue, L., Vishwanathan, S.A., Kilembe, W., et al. “Role Of Transmitted Gag CTL Polymorphisms In Defining Replicative Capacity And Early HIV-1 Pathogenesis.” PLoS Pathog. 2012;8(11):e1003041. doi: 10.1371/journal.ppat.1003041.

View Additional Publications

Continue reading

Wistar’s Medicinal Chemist Dr. Joe Salvino and the Journey to Drug Discovery

Joseph Salvino, Ph.D., medicinal chemist and professor in the Molecular & Cellular Oncogenesis Program and scientific director of the Molecular Screening & Protein Expression facility at The Wistar Institute, spent more than 20 years in the pharmaceutical industry’s drug discovery before coming to Wistar. Dr. Salvino collaborates with many Wistar scientists on programs to help identify novel small molecule lead compounds that could evolve into future drugs.

Here at the intersection of biology and chemistry is where Dr. Salvino and his team work best. Their medicinal and synthetic chemistry skills complement our investigators’ biology expertise. It’s a process wherein Dr. Salvino helps to optimize a hit compound our Wistar scientists identified or tries to identify a new lead compound for an interesting new target.

When Wistar scientists want to identify a compound that can produce a certain desired effect, Dr. Salvino works to optimize that compound’s ability to achieve its target effect. These early-stage compounds that show promise are called “hits,” and Dr. Salvino investigates these hits in a variety of biochemical settings.

Dr. Salvino’s expertise is in optimizing early-stage hits by improving target binding affinity and functional activity. His aim is to increase a compound’s biological potency and improve drug-like properties. To achieve this goal Dr. Salvino works closely with biologists to understand the molecular target. He focuses on how a small molecule will engage the target to elicit a biological response.

This crucial foundational research is the bedrock of the drug discovery process. It’s here that assays are developed with the throughput to support iterative medicinal chemistry optimization efforts that can quickly evaluate twenty or so compounds in a few days. The goal of lead optimization is to identify a suitable compound that could become a therapy to treat cancer and other disease. This is the compelling fundamental work that Wistar basic researchers accomplish before a drug discovery company considers translating what Wistar scientists have identified and potentially converts a Wistar discovery into a drug useful in health care.

As Wistar’s medicinal chemist, describe how you fit into Wistar’s scientific efforts?

I work in collaboration with Wistar scientists and scientists at neighboring universities to help identify a series of compounds suitable as a pharmacological means to modulate their target of interest. My job is to identify a suitable compound, part of a “hit-to-lead” series usually identified from a screening campaign, to test pharmacologically the effects of small molecule treatment both in vitro and in vivo.

In a lay friendly way tell us your process working with the scientists.

We work with other scientists by identifying and improving on small molecules that engage their protein target of interest. These small molecules may inhibit, stimulate, or degrade their protein and be biologically active in a cell expressing their protein, or where their protein is the cause for the disease we are trying to treat. My team needs to learn as much as we can about the molecular target from our collaborator.

We work with many Wistar investigators—typically those who are looking to identify or improve on a small molecule as a potential therapeutic agent for a disease related to their target. Often the investigator has already identified a small molecule to test their hypothesis. My team works in collaboration to improve or develop a new molecule, focusing on improving selectivity, potency, or its in vivo drug-like properties.

The Wistar Institute Molecular Screening & Protein Expression Core is under my direction. This group can develop assays that typically can be run in a plate-based format to provide a high-throughput approach to support our medicinal chemistry efforts. For example, when medicinal chemists are trying to identify an optimized compound, we need to synthesize and evaluate 10-50 different analogs that are related but have slight differences in their structure. We do this to probe for “structure activity relationships”—the changes required to improve binding affinity to a protein target or to improve its functional activity. Both binding affinity and functional efficacy are very important to optimize a molecule, even though its functional efficacy is what a biologist wants to study.

Interestingly, a typical drug discovery effort from a pharmaceutical company requires the synthesis of about 2000-3000 compounds per target to identify a development candidate.

How do you start working with scientists?

We start to work together because of a common interest in a target or a disease, such as treatment of melanoma, ovarian, or breast cancer or EBV associated cancer, or others. We normally start collaborating because of our common interests and complementary skills.

What aspects of your work do you like most?

I enjoy the interface between chemistry and biology. I love finding new compounds with interesting biological activity in collaboration with my colleagues, especially for interesting new targets. I like working with the screening core to help develop new methods to test compounds. We spend a lot of time synthesizing chemical probes, such as a binding probe, which greatly facilitate assay development. A binding probe, or also sometimes called a tracer, is used in a competitive binding assay, where an unlabeled compound will compete for binding with the tracer. For this type of study, we can determine the binding affinity of an unlabeled test compound.

Wistar does not make drugs or therapies but advances discoveries that can move into drug discovery as future therapeutics.