Fran Sverdrup, Ph.D.
Associate Professor
Epigenetic control of gene expression in facioscapulohumeral muscular dystrophy (FSHD) and the discovery of drugs to modulate this process.
Research Interests
My lab is focused on drug discovery in human genetic diseases as well as infectious diseases. We perform target identification and validation, drug screening, and preclinical evaluation of drug candidates. Our major current project targets facioscapulohumeral muscular dystrophy (FSHD), one of the most common forms of muscular dystrophy for which there is no treatment. Our goals are to identify druggable pathways that modulate expression of the toxic DUX4 gene that is responsible for FSHD and translate those finding into potential therapies.
Our approach is to perform high throughput screening of chemical libraries to identify compounds intended to epigenetically suppress the DUX4 gene. We evaluate new compounds by using biochemical and cell based assays to explore mechanisms and map pathways that lead to DUX4 expression. We are currently advancing three exciting classes of drugs that turn off DUX4 expression, including a robust lead optimization program that involves close collaboration with our medicinal chemistry colleagues. We are also utilizing a mouse xenograft model of human FSHD gene regulation to establish pharmacokinetic/pharmacodynamic (PK/PD) relationships of drug candidates. We have established a key collaboration with a pharmaceutical partner to advance one of these into human clinical trials.
A second interest is in anti-infectives research with recent programs targeting malaria, lymphatic filariasis, and African sleeping sickness. To accomplish these activities we maintain a network of collaborations with disease experts, medicinal chemists, pharmaceutical/biotech companies, and foundations.
Recent Publications
Discovery of an Orally Efficacious Pyrazolo[3,4-]pyrimidine Benzoxaborole as a Potent Inhibitor of
Discovery of an Orally Efficacious Pyrazolo[3,4-]pyrimidine Benzoxaborole as a Potent Inhibitor of
Cryptosporidiosis is a diarrheal disease caused by the parasite resulting in over 100,000 deaths annually. Here, we present a structure-activity relationship study of the benzoic acid position (R) of pyrazolo[3,4-]pyrimidine lead SLU-2815 (), an inhibitor of parasite phosphodiesterase PDE1, resulting in the discovery of benzoxaborole SLU-10906 () as a benzoic acid bioisostere. Benzoxaborole is 10-fold more potent than against the parasite in a cell-based infection model (EC = 0.19 μM) and non-cytotoxic. Furthermore, has a fast rate of parasite-killing and is orally efficacious in a mouse infection model (50 mg/kg BID), although relapse was observed 7 days post-drug treatment. The partial selectivity profile versus human phosphodiesterases is preserved with the benzoxaborole motif and represents an important feature to improve in future optimization. Benzoxaborole represents an important advance toward the optimization of the pyrazolo[3,4-]pyrimidine series and the identification of a drug to treat cryptosporidiosis.
SIX transcription factors are necessary for the activation of DUX4 expression in facioscapulohumeral muscular dystrophy
SIX transcription factors are necessary for the activation of DUX4 expression in facioscapulohumeral muscular dystrophy
Facioscapulohumeral muscular dystrophy (FSHD) is a common and progressive muscle wasting disease that is characterized by muscle weakness often first noticed in the face, the shoulder girdle and upper arms before progressing to the lower limb muscles. FSHD is caused by the misexpression of the Double Homeobox 4 (DUX4) transcription factor in skeletal muscle. While epigenetic derepression of D4Z4 macrosatellite repeats underlies DUX4 misexpression, our understanding of the complex transcriptional activation of DUX4 is incomplete.
Temporal variation in p38-mediated regulation of DUX4 in facioscapulohumeral muscular dystrophy
Temporal variation in p38-mediated regulation of DUX4 in facioscapulohumeral muscular dystrophy
Facioscapulohumeral muscular dystrophy (FSHD) is a degenerative muscle disease caused by loss of epigenetic silencing and ectopic reactivation of the embryonic double homeobox protein 4 gene (DUX4) in skeletal muscle. The p38 MAP kinase inhibitor losmapimod is currently being tested in FSHD clinical trials due to the finding that p38 inhibition suppresses DUX4 expression in preclinical models. However, the role of p38 in regulating DUX4 at different myogenic stages has not been investigated. We used genetic and pharmacologic tools in FSHD patient-derived myoblasts/myocytes to explore the temporal role of p38 in differentiation-induced DUX4 expression. Deletion of MAPK14/11 or inhibition of p38α/β caused a significant reduction in early differentiation-dependent increases in DUX4 and DUX4 target gene expression. However, in MAPK14/11 knockout cells, there remains a differentiation-associated increase in DUX4 and DUX4 target gene expression later in differentiation. Furthermore, pharmacologic inhibition of p38α/β only partially decreased DUX4 and DUX4 target gene expression in late differentiating myotubes. In xenograft studies, p38α/β inhibition by losmapimod failed to suppress DUX4 target gene expression in late FSHD xenografts. Our results show that while p38 is critical for DUX4 expression during early myogenesis, later in myogenesis a significant level of DUX4 expression is independent of p38α/β activity.
Structure-Activity Relationship Studies of the Aryl Acetamide Triazolopyridazines against Reveals Remarkable Role of Fluorine
Structure-Activity Relationship Studies of the Aryl Acetamide Triazolopyridazines against Reveals Remarkable Role of Fluorine
Our previous work identified compound (SLU-2633) as a potent lead compound toward the identification of a novel treatment for cryptosporidiosis, caused by the parasite (EC = 0.17 μM). While this compound is potent and orally efficacious, the mechanism of action and biological target(s) of this series are currently unknown. In this study, we synthesized 70 compounds to develop phenotypic structure-activity relationships around the aryl “tail” group. In this process, we found that 2-substituted compounds are inactive, confirmed that electron withdrawing groups are preferred over electron donating groups, and that fluorine plays a remarkable role in the potency of these compounds. The most potent compound resulting from this work is SLU-10482 (, EC = 0.07 μΜ), which was found to be orally efficacious with an ED < 5 mg/kg BID in a -infection mouse model, superior to SLU-2633.
Meeting report: the 2020 FSHD International Research Congress
Meeting report: the 2020 FSHD International Research Congress