Biomarker-focused multi-drug combination therapy and repurposing trial in mdx mice

Duchenne muscular dystrophy is initiated by dystrophin deficiency, but downstream pathophysiological pathways such as membrane instability, NFĸB activation, mitochondrial dysfunction, and induction of TGFβ fibrosis pathways are thought to drive the disability. Dystrophin replacement strategies are hopeful for addressing upstream dystrophin deficiency; however, all methods to date use semi-functional dystrophin proteins that are likely to trigger downstream pathways. Thus, combination therapies that can target multiple downstream pathways are important in treating DMD, even for dystrophin-replacement strategies. We sought to define blood pharmacodynamic biomarkers of drug response in the mdx mouse model of Duchenne muscular dystrophy using a series of repurposed drugs. Four-week-old mdx mice were treated for four weeks with four different drugs singly and in combination: vehicle, prednisolone, vamorolone, rituximab, β-aminoisobutyric acid (BAIBA) (11 treatment groups; n = 6/group). Blood was collected via cardiac puncture at study termination, and proteomic profiling was carried out using SOMAscan aptamer panels (1,310 proteins assayed). Prednisolone was tested alone and in combination with other drugs. It was found to have a good concordance of prednisolone-responsive biomarkers (56 increased by prednisolone, 39 decreased) focused on NFκB and TGFβ cascades. Vamorolone shared 45 (80%) of increased biomarkers and 13 (33%) of decreased biomarkers with prednisolone. Comparison of published human corticosteroid-responsive biomarkers to our mdx data showed 14% (3/22) concordance between mouse and human. Rituximab showed fewer drug-associated biomarkers, with the most significant being human IgG. On the other hand, BAIBA treatment (high and low dose) showed a drug-associated increase in 40 serum proteins and decreased 5 serum proteins. Our results suggest that a biomarker approach could be employed for assessing drug combinations in both mouse and human studies.

[1]  R. Finkel,et al.  Vamorolone trial in Duchenne muscular dystrophy shows dose-related improvement of muscle function , 2019, Neuromuscular Disorders.

[2]  Heeyoung Lee,et al.  Comparative Efficacy and Safety of Biosimilar Rituximab and Originator Rituximab in Rheumatoid Arthritis and Non-Hodgkin’s Lymphoma: A Systematic Review and Meta-analysis , 2019, BioDrugs.

[3]  E. Hoffman Pharmacotherapy of Duchenne Muscular Dystrophy. , 2019, Handbook of experimental pharmacology.

[4]  R. Inagi,et al.  SMPDL3b modulates insulin receptor signaling in diabetic kidney disease , 2019, Nature Communications.

[5]  T. Kuzel,et al.  The Discovery of Biomarkers in Cancer Immunotherapy , 2019, Computational and structural biotechnology journal.

[6]  E. Hoffman,et al.  Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy , 2019, Life Science Alliance.

[7]  Y. Horio,et al.  Resveratrol Ameliorates Mitophagy Disturbance and Improves Cardiac Pathophysiology of Dystrophin-deficient mdx Mice , 2018, Scientific Reports.

[8]  E. Hoffman,et al.  Serum biomarkers of glucocorticoid response and safety in anti-neutrophil cytoplasmic antibody-associated vasculitis and juvenile dermatomyositis , 2018, Steroids.

[9]  R. Finkel,et al.  Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first‐in‐class dissociative steroidal anti‐inflammatory drug , 2018, Pharmacological research.

[10]  E. Hoffman,et al.  Membrane Stabilization by Modified Steroid Offers a Potential Therapy for Muscular Dystrophy Due to Dysferlin Deficit , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

[11]  E. Hoffman,et al.  Muscle miRNAome shows suppression of chronic inflammatory miRNAs with both prednisone and vamorolone , 2018, Physiological genomics.

[12]  Ian Mitchelle S. de Vera,et al.  Cryptic glucocorticoid receptor-binding sites pervade genomic NF-κB response elements , 2018, Nature Communications.

[13]  Taeseung Lee,et al.  β-aminoisobutyric acid attenuates LPS-induced inflammation and insulin resistance in adipocytes through AMPK-mediated pathway , 2018, Journal of Biomedical Science.

[14]  E. Hoffman,et al.  Phase 1 trial of vamorolone, a first-in-class steroid, shows improvements in side effects via biomarkers bridged to clinical outcomes , 2018, Steroids.

[15]  M. Goddeeris,et al.  PPARδ modulation rescues mitochondrial fatty acid oxidation defects in the mdx model of muscular dystrophy. , 2018, Mitochondrion.

[16]  Ying Sun,et al.  Proteomic analysis reveals distinctive protein profiles involved in CD8+ T cell-mediated murine autoimmune cholangitis , 2018, Cellular & Molecular Immunology.

[17]  J. Assreuy,et al.  New Insights in Glucocorticoid Receptor Signaling—More Than Just a Ligand-Binding Receptor , 2017, Front. Endocrinol..

[18]  E. Hoffman,et al.  Identification of Pathway-Specific Serum Biomarkers of Response to Glucocorticoid and Infliximab Treatment in Children with Inflammatory Bowel Disease , 2016, Clinical and Translational Gastroenterology.

[19]  Kristy Brown,et al.  Serum pharmacodynamic biomarkers for chronic corticosteroid treatment of children , 2016, Scientific Reports.

[20]  E. Hoffman,et al.  Prednisone/prednisolone and deflazacort regimens in the CINRG Duchenne Natural History Study , 2015, Neurology.

[21]  E. Hoffman,et al.  Immune-mediated pathology in Duchenne muscular dystrophy , 2015, Science Translational Medicine.

[22]  Eric P. Hoffman,et al.  Large-scale serum protein biomarker discovery in Duchenne muscular dystrophy , 2015, Proceedings of the National Academy of Sciences.

[23]  Kristy Brown,et al.  Discovery of serum protein biomarkers in the mdx mouse model and cross-species comparison to Duchenne muscular dystrophy patients. , 2014, Human molecular genetics.

[24]  E. Hoffman,et al.  Asynchronous remodeling is a driver of failed regeneration in Duchenne muscular dystrophy , 2014, The Journal of cell biology.

[25]  Chad A. Cowan,et al.  β-Aminoisobutyric acid induces browning of white fat and hepatic β-oxidation and is inversely correlated with cardiometabolic risk factors. , 2014, Cell metabolism.

[26]  Kristy Brown,et al.  Sparing of the dystrophin-deficient cranial sartorius muscle is associated with classical and novel hypertrophy pathways in GRMD dogs. , 2013, The American journal of pathology.

[27]  E. Hoffman,et al.  VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects , 2013, EMBO molecular medicine.

[28]  E. Hoffman,et al.  The paradox of muscle hypertrophy in muscular dystrophy. , 2012, Physical medicine and rehabilitation clinics of North America.

[29]  Tracy R. Keeney,et al.  From SOMAmer-Based Biomarker Discovery to Diagnostic and Clinical Applications: A SOMAmer-Based, Streamlined Multiplex Proteomic Assay , 2011, PloS one.

[30]  M. Rastaldi,et al.  Rituximab Targets Podocytes in Recurrent Focal Segmental Glomerulosclerosis , 2011, Science Translational Medicine.

[31]  F. Bieber,et al.  Dystrophin analysis in duchenne muscular dystrophy: use in fetal diagnosis and in genetic counseling. , 1989, American journal of human genetics.