The Stegh Laboratory
for Convergence Brain Tumor Research
- …
The Stegh Laboratory
for Convergence Brain Tumor Research
- …
Nanotechnology
Metabolism
(Immune-)Metabolism
Me
Tumor Immunology
Early Phase Clinical Trials
MISSION
Our research program is aimed at understanding and therapeutically targeting the genetic program that underlies the pathogenesis of glioblastoma.
We apply a combination of cell/molecular biology, oncogenomic, and mouse engineering approaches, to understand fundamental mechanisms of metabolic adaptation and tumor-mediated immune suppression.
Known for our bench-to-bedside efforts to drive nanotechnologies toward clinical opportunity, we also develop novel nanotechnology-enabled approaches directed against actionable tumor and immune targets.
THE STEGH LABORATORY and THE BRAIN TUMOR CENTER at Washington University School of Medicine
Beginning in 2023, the Stegh laboratory will be housed within the new $616 million, 11-story and 609,000 square-foot, state-of-the-art Neuroscience Research Building. The building project is the largest in the history of Washington University’s School of Medicine and will span almost a block in the 200-acre Cortex Innovation Community — one of the fastest-growing business, innovation, and technology hubs in the United States. This infrastructure will enable and strengthen collaborations between academic research and industry partners, to rapidly translate fundamental science into advanced care for brain tumor patients.
KEY PUBLICATIONS
Development of Spherical Nucleic Acid Nano-Architectures
Spherical Nucleic Acids (SNAs) consists of a nanoparticle core densely functionalized with a shell of radially oriented synthetic oligonucleotides. The unique three-dimensional architecture of SNAs protects the oligonucleotides from nuclease-mediated degradation, increases oligonucleotide bioavailability, and in the absence of auxiliary transfection agents, enables robust uptake into tumor and immune cells through polyvalent association with cell surface pattern recognition receptors.
Nanotechnology enables new ways to diagnose and treat cancer. Nanomedicines can increase payload concentration at the disease site, reduce toxicity, and enhance therapeutic effect compared to drugs in their "free" form. SNAs emerged as a new class of oligonucleotide nanotherapeutics that are currently being explored for gene regulation, the activation of innate immunity, and the development of next-generation cancer vaccines.
Building on a decade long collaboration with Dr. Chad Mirkin's group at Northwestern University and a completed phase 0 clinical trial of gene-regualtory SNAs in patients with recurrent glioblastoma (NCT03020017), ongoing efforts in the laboratory focus on the development of next generation SNA architectures conjugated with gene-regulatory siRNA oligonucleotides, DNA oligionucleotides to activate innate DNA sensing pathways, and neoantigens for glioma vaccine development.
Defining Mechanisms of Metabolic Adaptation in Gliomas and the Glioma Tumor Microenvironment
Wild-type Isocitrate Dehydrogenases (IDHs) are enzymes that catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), NAD(P)H, and CO2. On molecular levels, IDHs epigenetically control gene expression through effects on α-KG-dependent dioxygenases, maintain redox balance, and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis. While gain-of-function mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effects, our studies and studies of other investigators identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down-regulated, as contributing to glioblastoma progression. Ongoing studies in the lab will answer the following questions:
1. How do wild-type IDHs control gene expression through chromatin and DNA modifications?
We will determine the extent of the epigenetic rewiring in wild-type IDH glioblastoma, the genes and pathways dysregulated, how these epigenetic changes support glioblastoma progression, and whether these pathways can be therapeutically targeted.
2. How do wild-type IDHs affect anti-tumor immunity?
We will determine the role of wild-type IDHs in shaping the systemic and the tumor-associated immune system, and will define the underlying mechanisms, in particular those mediated by α-KG and the ensuing effect on the regulation of immunogenic factors.
3. How does the IDH status impact progenitor cell transformation and glioma evolution?
We will determine whether and to what extent wild-type IDH1 expression in different progenitor cells (NSCs or NPCs versus OPCs) results in distinct tumor molecular and biologic properties and therapy susceptibility, using novel iPSC-derived models and inducible Cre alleles driven by NPCs or OPC-specific promoters to express or delete of wild-type IDHs in specific progenitor cell populations.
5. How can wild-type IDH1 be targeted?
We have identified small molecular inhibitors of wild-type IDH1. We will enroll these inhibitors into further preclinical testing, including detailed pharmacokinetic analysis, optimization of dosing, administration and formulation, comprehensive tumor regression analyses in advanced glioma mouse models and in vivo validation of its mechanism of action.
Defining Immunosuppression in Glioblastoma
Immunotherapies, including immune checkpoint inhibitors, can improve outcomes for patients with immunologically 'hot' tumors, but they have failed to provide survival benefits for patients with immunologically ‘cold’ tumors (i.e., low or non-T cell-inflamed), including those of glioblastoma. A limitation of using ICIs or other therapeutics to combat GBM immunosuppression is the low T cell effector response and high content of immunosuppressive myeloid cells. Our laboratory will determine precise mechanisms of immunosuppression and will identify actionable targets.
One focus in the laboratory is the study of the Activation of the Stimulator of Interferon Genes (STING) pathway, which represents one of the primary immune sensing mechanisms that bridges the innate and adaptive immune systems.
Tumor-derived double-stranded, but not single-stranded DNA is recognized by the DNA sensor cyclic GMP-AMP synthase (cGAS) in the cytosol of antigen-presenting cells. STING binds cyclic dinucleotides (CDNs), including 2′,3′–cyclic GMP-AMP (2′,3′-cGAMP), produced by cGAS in response to cytosolic DNA, and induces the expression of type I interferon and pro-inflammatory cytokines.
We will assess the importance of the cGAS-STING pathway for innate and adaptive immune responses against glioblastoma, will delineate novel mechanisms by which cGAS-activation triggers anti-tumor immunity, and will develop novel therapeutic approaches to engage this pathway, as monotherapy and when combined with other therapeutic modalities.
Developing unique Early Phase Clinical Trials for Nanomedicines
We conducted a first-in-human, window-of-opportunity phase 0 clinical trial of gene-regulatory SNAs (drug moniker: NU-0129) in patients with recurrent glioblastoma (NCT03020017), to assess SNA safety, pharmacokinetics, biodistribution, and target engagement. This clinical trial showed that intravenous infusion of SNAs, similar to results obtained in rodents and non-human primates, was safe at the dose administered and not associated with significant treatment-related toxicities. Mass spectrometry and X-ray fluorescence microscopy of resected glioblastoma patient tissue demonstrated that intravenously administered SNAs reached the patient tumor and accumulated in the cytoplasm of intraparenchymal tumor cells. SNA uptake into glioma cells was associated with a reduction in target protein expression and induction of apoptosis. These results establish SNAs as a safe, brain-penetrant precision medicine approach for the systemic delivery of siRNA oligonucleotides to intracranial tumor sites.
We are currently developing unique early phase clinical trial concepts to evaluate next-generation SNAs. In particular, we focus on the development of immunomodulatory SNAs, and the development of companion predictive and pharmacodynamic, imaging-based biomarkers.
SELECTED FUNDING
IN THE NEWS
Our graduate students....
Meet Akanksha Mahajan, a 5th year graduate student in the Stegh lab
Dr. Stegh inducted into the College of Fellows, American Institute for Medical and Biological Engineering
KEY COLLABORATORS
Chad A. Mirkin, PhD
Director of the International Institute for NanotechnologyRathmann Professor of ChemistryWeinberg College of Arts & Sciences. Professor of chemical and biological engineering, biomedical engineering, materials science & engineering, and medicine at Northwestern University.Development of Spherical Nucleic Acids nanoarchitectures
Daniel Wahl, MD, PhD
Associate ProfessorUniversity of MichiganDefining and targeting oncogenicity of wild-type IDH1
Sriram Venneti, MD
Al and Robert Glick Family Research Professor of PediatricsScientific Research DirectorCarr Pediatric Brain Tumor CenterJulie Taubman Reys Scholar in CancerAssociate Professor of PathologyAssociate Professor of PediatricsDefining and targeting oncogenicity of wild-type IDH1
Amy B. Heimberger, MD
Jean Malnati Miller ProfessorDirector of Research, Malnati Brain Tumor CenterProfessor of Neurological SurgeryDevelopment of cGAS-STING pathway agonists
Joshua Rubin, MD, PhD
Professor of PediatricsWashington University School of MedicineThe impact of sex differences on anti-glioma immunity
Priya U. Kumthekar MD
Associate Professor of NeurologyNorthwestern UniversityClinical translation of nano- medicines
Contact us
Alexander H. Stegh, PhD
Director of Research
Brain Tumor Center
Professor of Neurological Surgery
Washington University School of Medicine
Siteman Comprehensive Cancer Center
Address
Fort Labs
4730 Duncan St
Louis, MO 63110
Office: 6-112
ph: 314-747-1672
© 2019