Skip to main content

Interpretation, Integration, and Implementation of In Vitro Assay Data: The Predictive Toxicity Challenge

  • Protocol
  • First Online:
Drug-Induced Liver Toxicity

Part of the book series: Methods in Pharmacology and Toxicology ((MIPT))

Abstract

Human drug-induced liver injury (DILI) is still a leading cause of attrition in drug development and for adverse outcomes in the marketplace. Identifying DILI risk for humans in standard animal safety testing conducted before clinical trials, in advance of the drug regulatory review and approval process is sometimes elusive. Numerous mechanisms of small molecule driven hepatotoxicity have been elucidated (e.g., reactive metabolite formation, oxidative stress, mitochondrial inhibition, bile salt export pump inhibition) and are amenable to high-throughput screening approaches. It is possible to highlight and distinguish hazard gradations of risk for human DILI when these assays are consolidated into a single point of view. In some instances, these in vitro assays highlight the potential clinical hazard without animal safety studies confirming this view. Scientifically, the idea of developing a hazard matrix approach as a better option to animal studies requires a paradigm shift that challenges the status quo. Altering opinions and traditions to effect enduring change in this area require organizational effort and commitment. Utilizing a flexible, business-disciplined framework such as Accelerating Implementation Methodology (AIM), offers structure to implement organizational change. Common barriers associated with bringing lasting change are: inappropriate sponsorship (e.g., level, commitment, and influence), organizational commitment, lack of understanding of organizational culture, resistance to change, and not identifying key stakeholders, or not hearing stakeholders’ opposing views. Given time and appropriate scientific evidence, a hazard matrix approach to identifying compounds with higher potential to cause severe DILI in the clinic can be institutionalized within the pharmaceutical industry.

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 79.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 139.99
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. Kuhn TS (1970) The structure of scientific revolutions. International encyclopedia of unified science foundations of the unity of science, vol 2, no 2, 2nd edn. University of Chicago Press, Chicago

    Google Scholar 

  2. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER) (June 2012) Guidance for industry on: S2(R1) Genotoxicity testing and data interpretation for pharmaceuticals intended for human use. Available at: https://www.fda.gov/downloads/drugs/-guidancecomplianceregulatoryinformation/guidances/ucm074931.pdf

  3. Haasio K, Koponen A, Penttila KE, Nissinen E (2002) Effects of entacapone and tolcapone on mitochondrial membrane potential. Eur J Pharmacol 453(1):21–26

    Article  CAS  PubMed  Google Scholar 

  4. Yun GY, Kim SH, Kim SW, Joo JS, Kim JS, Lee ES, Lee BS, Kang SH, Moon HS, Sung JK, Lee HY, Kim KH (2016) Atypical onset of bicalutamide-induced liver injury. World J Gastroenterol 22(15):4062–4065. https://doi.org/10.3748/wjg.v22.i15.4062

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Greaves P, Williams A, Eve M (2004) First dose of potential new medicines to humans: how animals help. Nat Rev Drug Discov 3(3):226–236. https://doi.org/10.1038/nrd1329

    Article  CAS  PubMed  Google Scholar 

  6. Olson H, Betton G, Robinson D, Thomas K, Monro A, Kolaja G, Lilly P, Sanders J, Sipes G, Bracken W, Dorato M, Van Deun K, Smith P, Berger B, Heller A (2000) Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol 32(1):56–67. https://doi.org/10.1006/rtph.2000.1399

    Article  CAS  PubMed  Google Scholar 

  7. Laverty H, Benson C, Cartwright E, Cross M, Garland C, Hammond T, Holloway C, McMahon N, Milligan J, Park B, Pirmohamed M, Pollard C, Radford J, Roome N, Sager P, Singh S, Suter T, Suter W, Trafford A, Volders P, Wallis R, Weaver R, York M, Valentin J (2011) How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines? Br J Pharmacol 163(4):675–693. https://doi.org/10.1111/j.1476-5381.2011.01255.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, Pairaudeau G, Pennie WD, Pickett SD, Wang J, Wallace O, Weir A (2015) An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat Rev Drug Discov 14(7):475–486. https://doi.org/10.1038/nrd4609

    Article  CAS  PubMed  Google Scholar 

  9. Kaplowitz N (2005) Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 4(6):489–499. https://doi.org/10.1038/nrd1750

    Article  CAS  PubMed  Google Scholar 

  10. Lewis JH (2015) The art and science of diagnosing and managing drug-induced liver injury in 2015 and beyond. Clin Gastroenterol Hepatol 13(12):2173–2189.e8. https://doi.org/10.1016/j.cgh.2015.06.017

    Article  PubMed  Google Scholar 

  11. Dambach DM, Andrews BA, Moulin F (2005) New technologies and screening strategies for hepatotoxicity: use of in vitro models. Toxicol Pathol 33(1):17–26. https://doi.org/10.1080/01926230590522284

    Article  CAS  PubMed  Google Scholar 

  12. Gomez-Lechon MJ, Tolosa L, Donato MT (2016) Metabolic activation and drug-induced liver injury: in vitro approaches for the safety risk assessment of new drugs. J Appl Toxicol 36(6):752–768. https://doi.org/10.1002/jat.3277

    Article  CAS  PubMed  Google Scholar 

  13. Zhang J, Doshi U, Suzuki A, Chang CW, Borlak J, Li AP, Tong W (2016) Evaluation of multiple mechanism-based toxicity endpoints in primary cultured human hepatocytes for the identification of drugs with clinical hepatotoxicity: results from 152 marketed drugs with known liver injury profiles. Chem Biol Interact 255:3–11. https://doi.org/10.1016/j.cbi.2015.11.008

    Article  CAS  PubMed  Google Scholar 

  14. Nakayama S, Atsumi R, Takakusa H, Kobayashi Y, Kurihara A, Nagai Y, Nakai D, Okazaki O (2009) A zone classification system for risk assessment of idiosyncratic drug toxicity using daily dose and covalent binding. Drug Metab Dispos 37(9):1970–1977. https://doi.org/10.1124/dmd.109.027797

    Article  CAS  PubMed  Google Scholar 

  15. Sakatis MZ, Reese MJ, Harrell AW, Taylor MA, Baines IA, Chen L, Bloomer JC, Yang EY, Ellens HM, Ambroso JL, Lovatt CA, Ayrton AD, Clarke SE (2012) Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds. Chem Res Toxicol 25(10):2067–2082. https://doi.org/10.1021/tx300075j

    Article  CAS  PubMed  Google Scholar 

  16. Usui T, Mise M, Hashizume T, Yabuki M, Komuro S (2009) Evaluation of the potential for drug-induced liver injury based on in vitro covalent binding to human liver proteins. Drug Metab Dispos 37(12):2383–2392. https://doi.org/10.1124/dmd.109.028860

    Article  CAS  PubMed  Google Scholar 

  17. Park BK, Boobis A, Clarke S, Goldring CE, Jones D, Kenna JG, Lambert C, Laverty HG, Naisbitt DJ, Nelson S, Nicoll-Griffith DA, Obach RS, Routledge P, Smith DA, Tweedie DJ, Vermeulen N, Williams DP, Wilson ID, Baillie TA (2011) Managing the challenge of chemically reactive metabolites in drug development. Nat Rev Drug Discov 10(4):292–306. https://doi.org/10.1038/nrd3408

    Article  CAS  PubMed  Google Scholar 

  18. Thompson RA, Isin EM, Li Y, Weaver R, Weidolf L, Wilson I, Claesson A, Page K, Dolgos H, Kenna JG (2011) Risk assessment and mitigation strategies for reactive metabolites in drug discovery and development. Chem Biol Interact 192(1–2):65–71. https://doi.org/10.1016/j.cbi.2010.11.002

    Article  CAS  PubMed  Google Scholar 

  19. Thompson RA, Isin EM, Li Y, Weidolf L, Page K, Wilson I, Swallow S, Middleton B, Stahl S, Foster AJ, Dolgos H, Weaver R, Kenna JG (2012) In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs. Chem Res Toxicol 25(8):1616–1632. https://doi.org/10.1021/tx300091x

    Article  CAS  PubMed  Google Scholar 

  20. Dykens JA, Will Y (2007) The significance of mitochondrial toxicity testing in drug development. Drug Discov Today 12(17–18):777–785. https://doi.org/10.1016/j.drudis.2007.07.013

    Article  CAS  PubMed  Google Scholar 

  21. Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, Pangalos MN (2014) Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework. Nat Rev Drug Discov 13(6):419–431. https://doi.org/10.1038/nrd4309

    Article  CAS  PubMed  Google Scholar 

  22. Keisu M, Andersson TB (2010) Drug-induced liver injury in humans: the case of ximelagatran. Handb Exp Pharmacol 196:407–418. https://doi.org/10.1007/978-3-642-00663-0_13

    Article  CAS  Google Scholar 

  23. Kenna JG (2014) Current concepts in drug-induced bile salt export pump (BSEP) interference. Curr Protoc Toxicol 61:23.7.1–23.7.15. https://doi.org/10.1002/0471140856.tx2307s61

    Article  Google Scholar 

  24. Dawson S, Stahl S, Paul N, Barber J, Kenna JG (2012) In vitro inhibition of the bile salt export pump correlates with risk of cholestatic drug-induced liver injury in humans. Drug Metab Dispos 40(1):130–138. https://doi.org/10.1124/dmd.111.040758

    Article  CAS  PubMed  Google Scholar 

  25. Gustafsson F, Foster AJ, Sarda S, Bridgland-Taylor MH, Kenna JG (2014) A correlation between the in vitro drug toxicity of drugs to cell lines that express human P450s and their propensity to cause liver injury in humans. Toxicol Sci 137(1):189–211. https://doi.org/10.1093/toxsci/kft223

    Article  CAS  PubMed  Google Scholar 

  26. Galie N, Hoeper MM, Gibbs JS, Simonneau G (2011) Liver toxicity of sitaxentan in pulmonary arterial hypertension. Eur Respir J 37(2):475–476. https://doi.org/10.1183/09031936.00194810

    Article  CAS  PubMed  Google Scholar 

  27. Humbert M, Segal ES, Kiely DG, Carlsen J, Schwierin B, Hoeper MM (2007) Results of European post-marketing surveillance of bosentan in pulmonary hypertension. Eur Respir J 30(2):338–344. https://doi.org/10.1183/09031936.00138706

    Article  CAS  PubMed  Google Scholar 

  28. McGoon MD, Frost AE, Oudiz RJ, Badesch DB, Galie N, Olschewski H, McLaughlin VV, Gerber MJ, Dufton C, Despain DJ, Rubin LJ (2009) Ambrisentan therapy in patients with pulmonary arterial hypertension who discontinued bosentan or sitaxsentan due to liver function test abnormalities. Chest 135(1):122–129. https://doi.org/10.1378/chest.08-1028

    Article  CAS  PubMed  Google Scholar 

  29. Owen K, Cross DM, Derzi M, Horsley E, Stavros FL (2012) An overview of the preclinical toxicity and potential carcinogenicity of sitaxentan (Thelin®), a potent endothelin receptor antagonist developed for pulmonary arterial hypertension. Regul Toxicol Pharmacol 64(1):95–103. https://doi.org/10.1016/j.yrtph.2012.05.017

    Article  CAS  PubMed  Google Scholar 

  30. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER) (2001) Overall summary and evaluation of preclinical pharmacodynamics, toxicokinetics and toxicology. Available at: https://www.fda.gov/ohrms/dockets/ac/01/briefing/3775b2_07_PharmToxReview.htm

  31. Brand MD, Nicholls DG (2011) Assessing mitochondrial dysfunction in cells. Biochem J 435(2):297–312. https://doi.org/10.1042/BJ20110162

    Article  CAS  PubMed  Google Scholar 

  32. Kenna JG, Stahl SH, Eakins JA, Foster AJ, Andersson LC, Bergare J, Billger M, Elebring M, Elmore CS, Thompson RA (2015) Multiple compound-related adverse properties contribute to liver injury caused by endothelin receptor antagonists. J Pharmacol Exp Ther 352(2):281–290. https://doi.org/10.1124/jpet.114.220491

    Article  CAS  PubMed  Google Scholar 

  33. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER) (Feb 2012) Guidance for industry on: Drug interaction studies-study design, data analysis, implications for dosing, and labeling recommendations. Available at: https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm292362.pdf

  34. Chen M, Zhang J, Wang Y, Liu Z, Kelly R, Zhou G, Fang H, Borlak J, Tong W (2013) The liver toxicity knowledge base: a systems approach to a complex end point. Clin Pharmacol Ther 93(5):409–412. https://doi.org/10.1038/clpt.2013.16

    Article  CAS  PubMed  Google Scholar 

  35. Pfizer (Dec 2010) Pfizer stops clinical trials of Thelin® and initiates voluntary product withdrawal in the interest of patient safety. Available at: http://www.pfizer.com/news/press-release/press-release-detail/pfizer_stops_clinical_trials_of_thelin_and_initiates_voluntary_product_withdrawal_in_the_interest_of_patient_safety

  36. Aleo MD, Luo Y, Swiss R, Bonin PD, Potter DM, Will Y (2014) Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump. Hepatology 60(3):1015–1022. https://doi.org/10.1002/hep.27206

    Article  CAS  PubMed  Google Scholar 

  37. Shah F, Leung L, Barton HA, Will Y, Rodrigues AD, Greene N, Aleo MD (2015) Setting clinical exposure levels of concern for drug-induced liver injury (DILI) using mechanistic in vitro assays. Toxicol Sci 147(2):500–514. https://doi.org/10.1093/toxsci/kfv152

    Article  CAS  PubMed  Google Scholar 

  38. Xu JJ, Henstock PV, Dunn MC, Smith AR, Chabot JR, de Graaf D (2008) Cellular imaging predictions of clinical drug-induced liver injury. Toxicol Sci 105(1):97–105. https://doi.org/10.1093/toxsci/kfn109

    Article  CAS  PubMed  Google Scholar 

  39. Chen M, Vijay V, Shi Q, Liu Z, Fang H, Tong W (2011) FDA-approved drug labeling for the study of drug-induced liver injury. Drug Discov Today 16(15–16):697–703. https://doi.org/10.1016/j.drudis.2011.05.007

    Article  PubMed  Google Scholar 

  40. Chen M, Suzuki A, Thakkar S, Yu K, Hu C, Tong W (2016) DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans. Drug Discov Today 21(4):648–653. https://doi.org/10.1016/j.drudis.2016.02.015

    Article  CAS  PubMed  Google Scholar 

  41. Greene N, Fisk L, Naven RT, Note RR, Patel ML, Pelletier DJ (2010) Developing structure-activity relationships for the prediction of hepatotoxicity. Chem Res Toxicol 23(7):1215–1222. https://doi.org/10.1021/tx1000865

    Article  CAS  PubMed  Google Scholar 

  42. Cumming JG, Davis AM, Muresan S, Haeberlein M, Chen H (2013) Chemical predictive modelling to improve compound quality. Nat Rev Drug Discov 12(12):948–962. https://doi.org/10.1038/nrd4128

    Article  CAS  PubMed  Google Scholar 

  43. Implementation Management Associates (2016) Accelerating Implementation Methodology (AIM). Available at: http://www.imaworldwide.com/

  44. Choi S (2003) Nefazodone (Serzone) withdrawn because of hepatotoxicity. Can Med Assoc J 169(11):1187

    Google Scholar 

  45. Kakuda TN (2000) Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther 22(6):685–708. https://doi.org/10.1016/S0149-2918(00)90004-3

    Article  CAS  PubMed  Google Scholar 

  46. Ulrich RG, Bacon JA, Branstetter DG, Cramer CT, Funk GM, Hunt CE, Petrella DK, Sun EL (1995) Induction of a hepatic toxic syndrome in the Dutch-belted rabbit by a quinoxalinone anxiolytic. Toxicology 98(1–3):187–198

    Article  CAS  PubMed  Google Scholar 

  47. Xu D, Nishimura T, Nishimura S, Zhang H, Zheng M, Guo YY, Masek M, Michie SA, Glenn J, Peltz G (2014) Fialuridine induces acute liver failure in chimeric TK-NOG mice: a model for detecting hepatic drug toxicity prior to human testing. PLoS Med 11(4):e1001628. https://doi.org/10.1371/journal.pmed.1001628

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tennant BC, Baldwin BH, Graham LA, Ascenzi MA, Hornbuckle WE, Rowland PH, Tochkov IA, Yeager AE, Erb HN, Colacino JM, Lopez C, Engelhardt JA, Bowsher RR, Richardson FC, Lewis W, Cote PJ, Korba BE, Gerin JL (1998) Antiviral activity and toxicity of fialuridine in the woodchuck model of hepatitis B virus infection. Hepatology 28(1):179–191. https://doi.org/10.1002/hep.510280124

    Article  CAS  PubMed  Google Scholar 

  49. Krzyzewski S, Khetani SR, Barros S (2011) Assessing chronic toxicity of fialuridine in a micropatterned hepatocyte co-culture model (Abstract 2523). Toxicologist Suppl Toxicol Sci 120(2):540

    Google Scholar 

  50. Zimmerman HJ (1999) Hepatotoxicity: the adverse effects of drugs and other chemicals on the liver, 2nd edn. Lippincott Williams & Wilkins, Philadelphia, PA

    Google Scholar 

  51. Reuben A (2004) Hy’s law. Hepatology 39(2):574–578

    Article  PubMed  Google Scholar 

  52. Reuben A (2008) Hy’s law explained. Available at: http://www.documentshare.org/general-reference/hys-law-explained/

  53. U.S. Department of Health and Human Services, Food and Drug Adeministration, Center for Drug Evaluation and Research (CDER) (July 2009) Guidance for industry on: drug-induced liver injury: premarketing clinical evaluation. Available at: https://www.fda.gov/downloads/Drugs/.../guidances/UCM174090.pdf

  54. Temple R (2006) Hy’s law: predicting serious hepatotoxicity. Pharmacoepidemiol Drug Saf 15(4):241–243. https://doi.org/10.1002/pds.1211

    Article  PubMed  Google Scholar 

  55. Maddrey WC (2005) Drug-induced hepatotoxicity: 2005. J Clin Gastroenterol 39(4 Suppl 2):S83–S89

    Article  PubMed  Google Scholar 

  56. Benza RL, Mehta S, Keogh A, Lawrence EC, Oudiz RJ, Barst RJ (2007) Sitaxsentan treatment for patients with pulmonary arterial hypertension discontinuing bosentan. J Heart Lung Transplant 26(1):63–69. https://doi.org/10.1016/j.healun.2006.10.019

    Article  PubMed  Google Scholar 

  57. Don GW, Joseph F, Celermajer DS, Corte TJ (2012) Ironic case of hepatic dysfunction following the global withdrawal of sitaxentan. Intern Med J 42(12):1351–1354. https://doi.org/10.1111/imj.12007

    Article  CAS  PubMed  Google Scholar 

  58. Avigan MI (2014) DILI and drug development: a regulatory perspective. Semin Liver Dis 34(2):215–226. https://doi.org/10.1055/s-0034-1375961

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J. Gerry Kenna .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Light, D.S., Aleo, M.D., Kenna, J.G. (2018). Interpretation, Integration, and Implementation of In Vitro Assay Data: The Predictive Toxicity Challenge. In: Chen, M., Will, Y. (eds) Drug-Induced Liver Toxicity. Methods in Pharmacology and Toxicology. Humana, New York, NY. https://doi.org/10.1007/978-1-4939-7677-5_17

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7677-5_17

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-4939-7676-8

  • Online ISBN: 978-1-4939-7677-5

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics