Skip to main content

Designing an Orally Available Nontoxic p38 Inhibitor with a Fragment-Based Strategy

  • Protocol
  • First Online:
Fragment-Based Methods in Drug Discovery

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1289))

Abstract

The MAPK p38 became a focal point of inflammatory research when it was recognized that it played a key role in the production of the pro-inflammatory molecules TNF-alpha, IL-beta, and cyclooxygenase-2 (COX-2). The pharmaceutical industry devoted enormous efforts to creating p38 inhibitors, because blocking p38 had the potential of downregulating a group of pro-inflammatory mediators, and thus, one drug could have a cocktail effect. The market potential seemed to be clearly established (Bonafede et al., Clinicoecon Outcomes Res 6:381–388, 2014) with a multiplicity of TNF-alpha antibodies and a soluble receptor (Mewar and Wilson, Br J Pharmacol 162:785–791, 2011) already on the market, although the relationship between TNF-alpha production and p38 activation is a complicated two-way (Sabio and Davis, Semin Immunol 26:237–245, 2014) signal transduction process. With the discovery that activated p38 stabilizes (Mancini and Di Battista, Inflamm Res 60:1083–1092, 2011) COX-2 mRNA and upregulates expression of IL-beta (Bachstetter and Van Eldik, Aging Dis 1:199–211, 2010) probably in a similar manner, inhibiting p38 appeared to be a way of blocking TNF-alpha, COX-2, and IL-beta simultaneously. At Locus Pharmaceuticals we jumped on this opportunity, because we believed that our fragment-based drug discovery approach was ideally suited for making a potent small molecule p38 inhibitor that did not bind in the ATP site, but also had the solubility, lack of planarity, and low molecular weight required of a clinical candidate. Just to be clear, in our experience highly planar compounds often result in poor pharmacokinetics, because they tend to bind strongly to plasma proteins. At Locus we typically repeated assays by adding increasing amounts of plasma to check for potency degradation in the presence of blood. We found this to be a useful early indicator of pharmacokinetics and in vivo behavior. It became clear from our work and the work of others that binding to the ATP site resulted in nonspecific isoform toxicities, but binding in the adjacent allosteric DFG-site resulted in molecules that were too planar and too hydrophobic. Applying the computational method of Simulated Annealing of Chemical Potential (SACP) to this problem, we at Locus were able to come up with surprising fragment substitution patterns that led to potent non-ATP p38 inhibitors with the solubility and lack of planarity that resulted in potent in vivo efficacy in rodents with 33 % oral bioavailability. By using the simulations, we made only a small number of molecules and created a high quality clinical candidate. We also did extensive co-crystallography work, which demonstrated that the compounds bound in the mode predicted by the simulations. Unfortunately, all p38 programs ultimately shut down, because compelling evidence emerged that inhibiting p38 had no long-term clinical (Genovese, Arthritis Rheum 60:317–320, 2009) benefit. Devoting a large amount of limited resources to a target that ultimately turns out to be a mistake because it was not properly validated is a fatal error for a small company, and this is one of the reasons that Locus ultimately failed.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 89.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.00
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Bonafede M, Joseph GJ, Shah N, Princic N, Harrison DJ (2014) Cost of tumor necrosis factor blockers per patient with rheumatoid arthritis in a multistate Medicaid population. Clinicoecon Outcomes Res 6:381–388

    Article  PubMed Central  PubMed  Google Scholar 

  2. Mewar D, Wilson AG (2011) Treatment of rheumatoid arthritis with tumour necrosis factor inhibitors. Br J Pharmacol 162:785–791

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Sabio G, Davis RJ (2014) TNF and MAP kinase signalling pathways. Semin Immunol 26:237–245

    Article  CAS  PubMed  Google Scholar 

  4. Mancini AD, Di Battista JA (2011) The cardinal role of the phospholipase A(2)/cyclooxygenase-2/prostaglandin E synthase/prostaglandin E(2) (PCPP) axis in inflammostasis. Inflamm Res 60:1083–1092

    Article  CAS  PubMed  Google Scholar 

  5. Bachstetter AD, Van Eldik LJ (2010) The p38 MAP kinase family as regulators of proinflammatory cytokine production in degenerative diseases of the CNS. Aging Dis 1:199–211

    PubMed Central  PubMed  Google Scholar 

  6. Genovese MC (2009) Inhibition of p38: has the fat lady sung? Arthritis Rheum 60:317–320

    Article  PubMed  Google Scholar 

  7. Han J, Lee JD, Bibbs L, Ulevitch RJ (1994) A MAP kinase targeted by endotoxin and hyperosmolarity in mammalian cells. Science 265:808–811

    Article  CAS  PubMed  Google Scholar 

  8. Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, McNulty D, Blumenthal MJ, Heys JR, Landvatter SW et al (1994) A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature 372:739–746

    Article  CAS  PubMed  Google Scholar 

  9. Dumas J, Hatoum-Mokdad H, Sibley R, Riedl B, Scott WJ, Monahan MK, Lowinger TB, Brennan C, Natero R, Turner T, Johnson JS, Schoenleber R, Bhargava A, Wilhelm SM, Housley TJ, Ranges GE, Shrikhande A (2000) 1-Phenyl-5-pyrazolyl ureas: potent and selective p38 kinase inhibitors. Bioorg Med Chem Lett 10:2051–2054

    Article  CAS  PubMed  Google Scholar 

  10. Regan J, Pargellis CA, Cirillo PF, Gilmore T, Hickey ER, Peet GW, Proto A, Swinamer A, Moss N (2003) The kinetics of binding to p38MAP kinase by analogues of BIRB 796. Bioorg Med Chem Lett 13:3101–3104

    Article  CAS  PubMed  Google Scholar 

  11. Pargellis C, Tong L, Churchill L, Cirillo PF, Gilmore T, Graham AG, Grob PM, Hickey ER, Moss N, Pav S, Regan J (2002) Inhibition of p38 MAP kinase by utilizing a novel allosteric binding site. Nat Struct Biol 9:268–272

    Article  CAS  PubMed  Google Scholar 

  12. Long DL, Loeser RF (2010) p38gamma mitogen-activated protein kinase suppresses chondrocyte production of MMP-13 in response to catabolic stimulation. Osteoarthritis Cartilage 18:1203–1210

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Pogozelski AR, Geng T, Li P, Yin X, Lira VA, Zhang M, Chi JT, Yan Z (2009) p38gamma mitogen-activated protein kinase is a key regulator in skeletal muscle metabolic adaptation in mice. PLoS One 4:e7934

    Article  PubMed Central  PubMed  Google Scholar 

  14. Bradbury MW, Stump D, Guarnieri F, Berk PD (2011) Molecular modeling and functional confirmation of a predicted fatty acid binding site of mitochondrial aspartate aminotransferase. J Mol Biol 412:412–422

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Guarnieri F (2004) Computational protein probing to identify binding sites. US Patent No. 6,735,530. Issued 11 May 2004

    Google Scholar 

  16. Clark M, Guarnieri F, Shkurko I, Wiseman J (2006) Grand canonical Monte Carlo simulation of ligand-protein binding. J Chem Inf Model 46:231–242

    Article  CAS  PubMed  Google Scholar 

  17. Moore WR Jr (2005) Maximizing discovery efficiency with a computationally driven fragment approach. Curr Opin Drug Discov Devel 8:355–364

    CAS  PubMed  Google Scholar 

  18. Kulp JL III, Blumenthal SN, Wang Q, Bryan RL, Guarnieri F (2012) A fragment-based approach to the SAMPL3 challenge. J Comput Aided Mol Des 26:583–594

    Article  CAS  PubMed  Google Scholar 

  19. Kulp JL III, Kulp JL Jr, Pompliano DL, Guarnieri F (2011) Diverse fragment clustering and water exclusion identify protein hot spots. J Am Chem Soc 133:10740–10743

    Article  CAS  PubMed  Google Scholar 

  20. Guarnieri F, Mezei M (1996) Simulated annealing of chemical potential: a general procedure for locating bound waters. Application to the study of the differential hydration propensities of the major and minor grooves of DNA. J Am Chem Soc 118:8493–8494

    Article  CAS  Google Scholar 

  21. Michelotti EL, Moffett KK, Nguyen D, Kelly MJ, Shetty R, Chai X, Northrop K, Namboodiri V, Campbell B, Flynn GA, Fujimoto T, Hollinger FP, Bukhtiyarova M, Springman EB, Karpusas M (2005) Two classes of p38alpha MAP kinase inhibitors having a common diphenylether core but exhibiting divergent binding modes. Bioorg Med Chem Lett 15:5274–5279

    Article  CAS  PubMed  Google Scholar 

  22. Qin Z (2012) The use of THP-1 cells as a model for mimicking the function and regulation of monocytes and macrophages in the vasculature. Atherosclerosis 221:2–11

    Article  CAS  PubMed  Google Scholar 

  23. Oprea TI (2002) Current trends in lead discovery: are we looking for the appropriate properties? Mol Divers 5:199–208

    Article  PubMed  Google Scholar 

  24. Moffett K, Konteatis Z, Nguyen D, Shetty R, Ludington J, Fujimoto T, Lee KJ, Chai X, Namboodiri H, Karpusas M, Dorsey B, Guarnieri F, Bukhtiyarova M, Springman E, Michelotti E (2011) Discovery of a novel class of non-ATP site DFG-out state p38 inhibitors utilizing computationally assisted virtual fragment-based drug design (vFBDD). Bioorg Med Chem Lett 21:7155–7165

    Article  CAS  PubMed  Google Scholar 

  25. Zhou JS, Xing W, Friend DS, Austen KF, Katz HR (2007) Mast cell deficiency in Kit(W-sh) mice does not impair antibody-mediated arthritis. J Exp Med 204:2797–2802

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Cohen SB, Cheng TT, Chindalore V, Damjanov N, Burgos-Vargas R, Delora P, Zimany K, Travers H, Caulfield JP (2009) Evaluation of the efficacy and safety of pamapimod, a p38 MAP kinase inhibitor, in a double-blind, methotrexate-controlled study of patients with active rheumatoid arthritis. Arthritis Rheum 60:335–344

    Article  CAS  PubMed  Google Scholar 

  27. Stroes E, Colquhoun D, Sullivan D, Civeira F, Rosenson RS, Watts GF, Bruckert E, Cho L, Dent R, Knusel B, Xue A, Scott R, Wasserman SM, Rocco M (2014) Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol 63:2541–2548

    Article  CAS  PubMed  Google Scholar 

  28. Robinson JG, Nedergaard BS, Rogers WJ, Fialkow J, Neutel JM, Ramstad D, Somaratne R, Legg JC, Nelson P, Scott R, Wasserman SM, Weiss R (2014) Effect of evolocumab or ezetimibe added to moderate- or high-intensity statin therapy on LDL-C lowering in patients with hypercholesterolemia: the LAPLACE-2 randomized clinical trial. JAMA 311:1870–1882

    Article  CAS  PubMed  Google Scholar 

  29. Koren MJ, Lundqvist P, Bolognese M, Neutel JM, Monsalvo ML, Yang J, Kim JB, Scott R, Wasserman SM, Bays H (2014) Anti-PCSK9 Monotherapy for Hypercholesterolemia: The MENDEL-2 Randomized, Controlled Phase III Clinical Trial of Evolocumab. J Am Coll Cardiol 63:2531–2540

    Article  CAS  PubMed  Google Scholar 

  30. Blom DJ, Hala T, Bolognese M, Lillestol MJ, Toth PD, Burgess L, Ceska R, Roth E, Koren MJ, Ballantyne CM, Monsalvo ML, Tsirtsonis K, Kim JB, Scott R, Wasserman SM, Stein EA (2014) A 52-week placebo-controlled trial of evolocumab in hyperlipidemia. N Engl J Med 370:1809–1819

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Frank Guarnieri .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer Science+Business Media New York

About this protocol

Cite this protocol

Guarnieri, F. (2015). Designing an Orally Available Nontoxic p38 Inhibitor with a Fragment-Based Strategy. In: Klon, A. (eds) Fragment-Based Methods in Drug Discovery. Methods in Molecular Biology, vol 1289. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-2486-8_15

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-2486-8_15

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-2485-1

  • Online ISBN: 978-1-4939-2486-8

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics