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

Michael Ziemba, Molly Barkhouse, Kitipong Uaesoontrachoon, Mamta Giri, Yetrib Hathout, Utkarsh J Dang, Heather Gordish-Dressman, Kanneboyina Nagaraju, Eric P Hoffman, Michael Ziemba, Molly Barkhouse, Kitipong Uaesoontrachoon, Mamta Giri, Yetrib Hathout, Utkarsh J Dang, Heather Gordish-Dressman, Kanneboyina Nagaraju, Eric P Hoffman

Abstract

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.

Conflict of interest statement

Declaration of Interest: KN and EPH hold founder shares and management positions in AGADA Biosciences. MB and KU are employees of AGADA Biosciences. AGADA Biosciences is a contract research organization that received a competitive grant from Foundation to Eradicate Duchenne to perform studies presented in this paper. UD receives consultancy funds from ReveraGen BioPharma. KN and EPH hold founder shares and management positions in ReveraGen BioPharma, the sponsor of vamorolone, a drug in clinical development for Duchenne muscular dystrophy. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Ingenuity Pathway Analysis of prednisolone-responsive…
Fig 1. Ingenuity Pathway Analysis of prednisolone-responsive biomarkers identifies inflammatory cytokine and NFκB pathways shared with vamorolone.
The two most significant enrichment of protein-protein interactions for prednisolone-responsive biomarkers are shown (left panels), with vamorolone-responsive proteins overlaid onto the prednisolone networks. Red indicates upregulation, and green indicates downregulation, and color intensity correlates with the size of expression change. The multicolored elements are protein complexes where the underlying data and documentation are inconsistent with IPAs search algorithms, and therefore, the details are not shown.
Fig 2. Ingenuity Pathway Analysis of BAIBA-responsive…
Fig 2. Ingenuity Pathway Analysis of BAIBA-responsive biomarkers identifies modulation of inflammatory pathways.
The top IPA network identified with high dose BAIBA treatment data is shown.

References

    1. Bieber FR, Hoffman EP, Amos JA. Dystrophin analysis in Duchenne muscular dystrophy: Use in fetal diagnosis and in genetic counseling. Am J Hum Genet. 1989;45: 362–367.
    1. Kornegay JN, Childers MK, Bogan DJ, Bogan JR, Nghiem P, Wang J, et al. The Paradox of Muscle Hypertrophy in Muscular Dystrophy. 2018;23: 1–29. 10.1016/j.pmr.2011.11.014.The
    1. Nghiem PP, Hoffman EP, Mittal P, Brown KJ, Schatzberg SJ, Ghimbovschi S, et al. Sparing of the dystrophin-deficient cranial sartorius muscle is associated with classical and novel hypertrophy pathways in GRMD dogs. Am J Pathol. 2013;183: 1411–1424. 10.1016/j.ajpath.2013.07.013
    1. Dadgar S, Wang Z, Johnston H, Kesari A, Nagaraju K, Chen YW, et al. Asynchronous remodeling is a driver of failed regeneration in Duchenne muscular dystrophy. J Cell Biol. 2014;207: 139–158. 10.1083/jcb.201402079
    1. Bello L, Gordish- H, Morgenroth LP, Henricson EK, Duong T, Hoffman EP, et al. Prednisone / prednisolone and deflazacort regimens in the CINRG Duchenne Natural History Study. Neurology. 2015;85: 1048–1055. 10.1212/WNL.0000000000001950
    1. Conklin ALS, Damsker JM, Hoffman EP, Jusko WJ, Mah JK, Guglieri M, et al. Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first- in-class dissociative steroidal anti-inflammatory drug. 2012;136: 140–150. 10.1016/j.phrs.2018.09.007
    1. Hoffman EP, Riddle V, Siegler MA, Dickerson D, Backonja M, Kramer WG, et al. Phase 1 trial of vamorolone, a first-in-class steroid, shows improvements in side effects via biomarkers bridged to clinical outcomes. Steroids. 2018;134: 43–52. 10.1016/j.steroids.2018.02.010
    1. Hoffman EP, Schwartz BD, Mengle-Gaw LJ, Smith EC, Castro D, Mah JK, et al. Vamorolone trial in Duchenne muscular dystrophy shows dose-related improvement of muscle function. Neurology. 2019;93: E1312–E1323. 10.1212/WNL.0000000000008168
    1. Sreetama SC, Chandra G, Van der Meulen JH, Ahmad MM, Suzuki P, Bhuvanendran S, et al. Membrane Stabilization by Modified Steroid Offers a Potential Therapy for Muscular Dystrophy Due to Dysferlin Deficit. Mol Ther. 2018;26: 2231–2242. 10.1016/j.ymthe.2018.07.021
    1. Kuno A, Hosoda R, Sebori R, Hayashi T, Sakuragi H, Tanabe M, et al. Resveratrol Ameliorates Mitophagy Disturbance and Improves Cardiac Pathophysiology of Dystrophin-deficient mdx Mice. Sci Rep. 2018;8: 1–12. 10.1038/s41598-017-17765-5
    1. Bell EL, Shine RW, Dwyer P, Olson L, Truong J, Fredenburg R, et al. PPARδ modulation rescues mitochondrial fatty acid oxidation defects in the mdx model of muscular dystrophy. Mitochondrion. 2019;46: 51–58. 10.1016/j.mito.2018.02.006
    1. Roberts LD, Boström P, O’Sullivan JF, Schinzel RT, Lewis GD, Dejam A, et al. β-Aminoisobutyric acid induces browning of white fat and hepatic β-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 2014;19: 96–108. 10.1016/j.cmet.2013.12.003
    1. Jung TW, Park HS, Choi GH, Kim D, Lee T. β-aminoisobutyric acid attenuates LPS-induced inflammation and insulin resistance in adipocytes through AMPK-mediated pathway. J Biomed Sci. 2018;25: 1–9. 10.1186/s12929-017-0402-4
    1. George AP, Kuzel TM, Zhang Y, Zhang B. The Discovery of Biomarkers in Cancer Immunotherapy. Comput Struct Biotechnol J. 2019;17: 484–497. 10.1016/j.csbj.2019.03.015
    1. Hathout Y, Brody E, Clemens PR, Cripe L, Delisle RK, Furlong P, et al. Large-scale serum protein biomarker discovery in Duchenne muscular dystrophy. Sci Rep. 2016;6:31727 10.1038/srep31727
    1. Hathout Y, Conklin LS, Seol H, Gordish-dressman H, Brown KJ, Morgenroth LP, et al. Serum pharmacodynamic biomarkers for chronic corticosteroid treatment of children. Sci Rep. 2016;6, 31727: 1–10. 10.1038/srep31727
    1. Conklin LS, Merkel PA, Pachmand LM, Parikhe H, Tawalbehf S, Damsker JM, et al. Serum biomarkers of glucocorticoid response and safety in anti-neutrophil cytoplasmic antibody-associated vasculitis and juvenile dermatomyositis Laurie S. Conklin. 2018; 1–28.
    1. Rosenberg AS, Puig M, Nagaraju K, Hoffman EP, Villalta SA, Rao VA, et al. Immune-mediated pathology in Duchenne muscular dystrophy. Science Translational Medicine. 2015. 10.1126/scitranslmed.aaa7322
    1. Hoffman EP. Pharmacotherapy of Duchenne Muscular Dystrophy. Handb Exp Pharmacol. 2019. p. 10.1007/164_2019_256 Aug 3. 2019
    1. Scheschowitsch K, Leite JA, Assreuy J. New insights in glucocorticoid receptor signaling-more than just a ligand-binding receptor. Front Endocrinol (Lausanne). 2017;8: 19–21. 10.3389/fendo.2017.00016
    1. Heier CR, Damsker JM, Yu Q, Dillingham BC, Huynh T, Van der Meulen JH, et al. VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects. EMBO Mol Med. 2013;5: 1569–1585. 10.1002/emmm.201302621
    1. Heier CR, Yu Q, Fiorillo AA, Tully CB, Tucker A, Mazala DA, et al. Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy. Life Sci alliance. 2019;2: 1–16. 10.26508/lsa.201800186
    1. Fiorillo AA, Tully CB, Damsker JM, Nagaraju K, Hoffman EP, Heier CR. Muscle miRNAome shows suppression of chronic inflammatory miRNAs with both prednisone and vamorolone. Physiol Genomics. 2018;50: 735–745. 10.1152/physiolgenomics.00134.2017
    1. lee S, Lee H, Kim E. Comparative Efficacy and Safety of Biosimilar Rituximab and Originator Rituximab in Rheumatoid Arthritis and Non-Hodgkin’s Lymphoma: A Systematic Review and Meta-analysis. BioDrugs.: 469–483. 10.1007/s40259-019-00376-z
    1. Fornoni A, Sageshima J, Wei C, Merscher-Gomez S, Aguillon-Prada R, Jauregui AN, et al. Rituximab targets podocytes in recurrent focal segmental glomerulosclerosis. Sci Transl Med. 2011;3: 1–21. 10.1126/scitranslmed.3002231
    1. SomaLogic. SOMAscan Quality Statement. 2016.
    1. Heier CR, Fiorillo AA, Chaisson E, Gordish-dressman H, Hathout Y, Damsker JM, et al. Identification of Pathway-Specific Serum Biomarkers of Response to Glucocorticoid and Infliximab Treatment in Children with Inflammatory Bowel Disease. Nat Publ Gr. 2016; 1–8. 10.1038/ctg.2016.49
    1. Mitrofanova A, Mallela SK, Ducasa GM, Yoo TH, Rosenfeld-Gur E, Zelnik ID, et al. SMPDL3b modulates insulin receptor signaling in diabetic kidney disease. Nat Commun. 2019;10 10.1038/s41467-018-07709-6
    1. Kraemer S, Vaught JD, Bock C, Gold L, Katilius E, Keeney TR, et al. From SOMAmer-based biomarker discovery to diagnostic and clinical applications: A SOMAmer-based, streamlined multiplex proteomic assay. PLoS One. 2011;6 10.1371/journal.pone.0026332
    1. Zhang W, Zhang R, Zhang J, Sun Y, Leung PS, Yang GX, et al. Proteomic analysis reveals distinctive protein profiles involved in CD8+ T cell-mediated murine autoimmune cholangitis. Cell Mol Immunol. 2018;15: 756–767. 10.1038/cmi.2017.149
    1. Hathout Y, Marathi RL, Rayavarapu S, Zhang A, Brown KJ, Seol H, et al. Discovery of serum protein biomarkers in the mdx mouse model and cross-species comparison to Duchenne muscular dystrophy patients. Hum Mol Genet. 2014;23: 6458–6469. 10.1093/hmg/ddu366
    1. Hudson WH, Vera IMSD, Nwachukwu JC, Weikum ER, Herbst AG, Yang Q, et al. Cryptic glucocorticoid receptor-binding sites pervade genomic NF-κB response elements. Nat Commun. 2018;9 10.1038/s41467-018-03780-1

Source: PubMed

3
S'abonner