A new MAPK13-guided inhibitor for respiratory inflammation and mucus production

Common respiratory diseases continue to represent a major public health problem, and much of the morbidity and mortality is due to airway inflammation and mucus production. Our previous work identified a mitogen-activated protein kinase (MAPK) known as MAPK13 that is activated in airway disease and is required for mucus production in human cell-culture models. However, only weak first-generation MAPK13 inhibitors were made to confirm gene-knockdown function, and there was no extension to effectiveness in vivo. Here we report the discovery of a first-in-class MAPK13 inhibitor (designated NuP-3) that down-regulates type-2 cytokine-stimulated mucus production in air-liquid interface and organoid cultures of human airway epithelial cells. We also show that NuP-3 treatment effectively attenuates respiratory inflammation and mucus production in new minipig models of airway disease after type-2 cytokine challenge or respiratory viral infection. Treatment also down-regulates biomarkers linked to basal-epithelial stem cell activation as an upstream site for target engagement. The results thereby provide proof-of-concept for a novel small-molecule kinase inhibitor to modify as yet uncorrected features of respiratory airway disease including stem cell reprogramming towards inflammation and mucus production.

[1]  M. Holtzman,et al.  Lung Remodeling Regions in Long-Term Coronavirus Disease 2019 Feature Basal Epithelial Cell Reprogramming , 2023, The American journal of pathology.

[2]  Shamus P. Keeler,et al.  Chloride channel accessory 1 (CLCA1) gene deficiency causes selective loss of mucus production in a new pig model. , 2022, American journal of physiology. Lung cellular and molecular physiology.

[3]  Samantha A. Morris,et al.  Basal-epithelial stem cells cross an alarmin checkpoint for post-viral lung disease. , 2021, The Journal of clinical investigation.

[4]  Shamus P. Keeler,et al.  TLR3-Activated Monocyte-Derived Dendritic Cells Trigger Progression from Acute Viral Infection to Chronic Disease in the Lung , 2021, The Journal of Immunology.

[5]  Benjamin Bowe,et al.  High-dimensional characterization of post-acute sequelae of COVID-19 , 2021, Nature.

[6]  Shamus P. Keeler,et al.  Group 2 Innate Lymphoid Cells Must Partner with the Myeloid–Macrophage Lineage for Long-Term Postviral Lung Disease , 2020, The Journal of Immunology.

[7]  A. Saqi,et al.  Distinct stem/progenitor cells proliferate to regenerate the trachea, intrapulmonary airways and alveoli in COVID-19 patients , 2020, Cell Research.

[8]  Matthew Rowland,et al.  Trials of anti-tumour necrosis factor therapy for COVID-19 are urgently needed , 2020, The Lancet.

[9]  S. Laufer,et al.  An updated patent review of p38 MAP kinase inhibitors (2014-2019) , 2020, Expert opinion on therapeutic patents.

[10]  Shamus P. Keeler,et al.  Respiratory Enterovirus (like Parainfluenza Virus) Can Cause Chronic Lung Disease if Protection by Airway Epithelial STAT1 Is Lost , 2019, The Journal of Immunology.

[11]  Shamus P. Keeler,et al.  Influenza A Virus Infection Causes Chronic Lung Disease Linked to Sites of Active Viral RNA Remnants , 2018, The Journal of Immunology.

[12]  Si-Youn Song,et al.  High Concentration of Insulin Induces MUC5AC Expression via Phosphoinositide 3 Kinase/AKT and Mitogen-activated Protein Kinase Signaling Pathways in Human Airway Epithelial Cells , 2018, American journal of rhinology & allergy.

[13]  E. Hoffman,et al.  Mucus plugs in patients with asthma linked to eosinophilia and airflow obstruction , 2018, The Journal of clinical investigation.

[14]  Heiner Koch,et al.  The target landscape of clinical kinase drugs , 2017, Science.

[15]  Jiachun Lu,et al.  MicroRNA-218 acts by repressing TNFR1-mediated activation of NF-κB, which is involved in MUC5AC hyper-production and inflammation in smoking-induced bronchiolitis of COPD. , 2017, Toxicology letters.

[16]  E. Hoffman,et al.  Airway Mucin Concentration as a Marker of Chronic Bronchitis , 2017, The New England journal of medicine.

[17]  A. Risco,et al.  Mitogen Activated Protein Kinases ( MAPKs ) , New Stars in the MAPK Galaxy , 2016 .

[18]  K. Devarajan,et al.  Kinase Inhibitor Profiling Reveals Unexpected Opportunities to Inhibit Disease-Associated Mutant Kinases. , 2016, Cell reports.

[19]  John P. Overington,et al.  Comprehensive characterization of the Published Kinase Inhibitor Set , 2016, Nature Biotechnology.

[20]  Emilio Benfenati,et al.  Identification of structural alerts for liver and kidney toxicity using repeated dose toxicity data , 2015, Chemistry Central Journal.

[21]  Li Xing,et al.  Kinase hinge binding scaffolds and their hydrogen bond patterns. , 2015, Bioorganic & medicinal chemistry.

[22]  M. D. Hill,et al.  Applications of Fluorine in Medicinal Chemistry. , 2015, Journal of medicinal chemistry.

[23]  M. Colonna,et al.  TREM-2 promotes macrophage survival and lung disease after respiratory viral infection , 2015, The Journal of experimental medicine.

[24]  R. Pierce,et al.  Long-term IL-33-producing epithelial progenitor cells in chronic obstructive lung disease. , 2013, The Journal of clinical investigation.

[25]  W. MacNee,et al.  Efficacy and safety of the oral p38 inhibitor PH-797804 in chronic obstructive pulmonary disease: a randomised clinical trial , 2013, Thorax.

[26]  M. Holtzman,et al.  IL-13-induced airway mucus production is attenuated by MAPK13 inhibition. , 2012, The Journal of clinical investigation.

[27]  S. Laufer,et al.  Targeting the hinge glycine flip and the activation loop: novel approach to potent p38α inhibitors. , 2012, Journal of medicinal chemistry.

[28]  D. Hill,et al.  A Periciliary Brush Promotes the Lung Health by Separating the Mucus Layer from Airway Epithelia , 2012, Science.

[29]  Satoko Takizawa,et al.  A possible mechanism for hepatotoxicity induced by BIRB‐796, an orally active p38 mitogen‐activated protein kinase inhibitor , 2011, Journal of applied toxicology : JAT.

[30]  Dean M. Messing,et al.  Discovery of PH-797804, a highly selective and potent inhibitor of p38 MAP kinase. , 2011, Bioorganic & medicinal chemistry letters.

[31]  A. Kuglstatter,et al.  X-ray crystal structure of JNK2 complexed with the p38alpha inhibitor BIRB796: insights into the rational design of DFG-out binding MAP kinase inhibitors. , 2010, Bioorganic & medicinal chemistry letters.

[32]  R. Adams,et al.  Global Initiative for Chronic Obstructive Lung Disease stage 0 is associated with excess FEV(1) decline in a representative population sample. , 2010, Chest.

[33]  Michael Shaw,et al.  Clinical aspects of pandemic 2009 influenza A (H1N1) virus infection. , 2010, The New England journal of medicine.

[34]  J. McQualter,et al.  Evidence of an epithelial stem/progenitor cell hierarchy in the adult mouse lung , 2010, Proceedings of the National Academy of Sciences.

[35]  M. Holtzman,et al.  The role of CLCA proteins in inflammatory airway disease. , 2009, Annual review of physiology.

[36]  Charles Schmidt An evolving threat , 2008, Nature Medicine.

[37]  Jeffrey D. Morton,et al.  Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease , 2008, Nature Medicine.

[38]  L. Fabbri,et al.  Increased activation of p38 MAPK in COPD , 2008, European Respiratory Journal.

[39]  P. Paré,et al.  Survival after lung volume reduction in chronic obstructive pulmonary disease: insights from small airway pathology. , 2007, American journal of respiratory and critical care medicine.

[40]  Jeffrey D. Morton,et al.  Blocking airway mucous cell metaplasia by inhibiting EGFR antiapoptosis and IL-13 transdifferentiation signals. , 2006, The Journal of clinical investigation.

[41]  S. Akira,et al.  Pathogen recognition with Toll-like receptors. , 2005, Current opinion in immunology.

[42]  H. H. Choo,et al.  Inhaled p38alpha mitogen-activated protein kinase antisense oligonucleotide attenuates asthma in mice. , 2005, American journal of respiratory and critical care medicine.

[43]  P. Paré,et al.  The nature of small-airway obstruction in chronic obstructive pulmonary disease. , 2004, The New England journal of medicine.

[44]  C. Pargellis,et al.  The kinetics of binding to p38MAP kinase by analogues of BIRB 796. , 2003, Bioorganic & medicinal chemistry letters.

[45]  P. Paré,et al.  Characterization of airway plugging in fatal asthma. , 2003, The American journal of medicine.

[46]  Stephen R. Johnson,et al.  Molecular properties that influence the oral bioavailability of drug candidates. , 2002, Journal of medicinal chemistry.

[47]  L. Tong,et al.  Inhibition of p38 MAP kinase by utilizing a novel allosteric binding site , 2002, Nature Structural Biology.

[48]  A. Nicholson,et al.  Expression of respiratory mucins in fatal status asthmaticus and mild asthma , 2002, Histopathology.

[49]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[50]  R R Osborn,et al.  SB 239063, a potent p38 MAP kinase inhibitor, reduces inflammatory cytokine production, airways eosinophil infiltration, and persistence. , 2000, The Journal of pharmacology and experimental therapeutics.

[51]  J. Lechner,et al.  A serum-free method for culturing normal human bronchial epithelial cells at clonal density , 1985 .

[52]  A. Buckpitt,et al.  Evidence for hepatic formation, export and covalent binding of reactive naphthalene metabolites in extrahepatic tissues in vivo. , 1983, The Journal of pharmacology and experimental therapeutics.

[53]  J. Pouysségur,et al.  A novel role for IkappaB kinase (IKK) alpha and IKKbeta in ERK-dependent up-regulation of MUC5AC mucin transcription by Streptococcus pneumoniae. , 2007, Journal of immunology.

[54]  M. Homma,et al.  Sendai virus. , 1978, Advances in virus research.

[55]  Tsyvkin Mv,et al.  LEADING CAUSES OF DEATH. , 1964, WHO chronicle.