Investigative safety strategies to improve success in drug development

Authors

    Franck Atienzar, Annie Delaunois, Frédéric Brouta, Miranda Cornet, Renaud Fleurance, Helga Gerets, Stéphanie Glineur, Catrin Hasselgren, Andrea Kiessling, Andre Nogueira da Costa, Marie-Luce Rosseels, Karen Tilmant, Jean-Pierre Valentin

DOI:

https://doi.org/10.18063/jmds.v2i1.139

Keywords:

safety attrition, drug development, target organ strategies, on and off target effects, hazard identification, risk assessment, mitigation plans

Abstract

Understanding and reducing attrition rate remains a key challenge in drug development. Preclinical and clinical safety issues still represent about 40% of drug discontinuation, of which cardiac and liver toxicities are the leading reasons. Reducing attrition rate can be achieved by various means, starting with a comprehensive evaluation of the potential safety issues associated to the primary target followed by an evaluation of undesirable secondary targets. To address these risks, a risk mitigation plan should be built at very early development stages, using a panel of in silico, in vitro, and in vivo models. While most pharmaceutical companies have developed robust safety strategies to de-risk genotoxicity and cardiotoxicity issues, partly driven by regulatory requirements; safety issues affecting other organs or systems, such as the central nervous system, liver, kidney, or gastro-intestinal system are less commonly addressed during early drug development. This paper proposes some de-risking strategies that can be applied to these target organ systems, including the use of novel biomarkers that can be easily integrated in both preclinical and clinical studies. Experiments to understand the mechanisms’ underlying toxicity are also important. Two examples are provided to demonstrate how such mechanistic studies can impact drug development. Novel trends in investigative safety are reviewed, such as computational modeling, mitochondrial toxicity assessment, and imaging technologies. Ultimately, understanding the predictive value of non-clinical safety testing and its translatability to humans will enable to optimize assays in order to address the key objectives of the drug discovery process, i.e., hazard identification, risk assessment, and mitigation 

References

Report to the President on propelling innovation in drug discovery, development, and evaluation, 2012, President’s Council of Advisors on Science and Technology (PCAST), viewed April 17, 2016,

Pammolli F, Magazzini L and Riccaboni M, 2011, The productivity crisis in pharmaceutical R&D. Nature Reviews Drug Discovery, vol.10(6): 428–438.

http://dx.doi.org/10.1038/nrd3405

Booth B and Zemmel R, 2004, Prospects for productivity. Nature Reviews Drug Discovery, vol.3(5): 451–456. http://dx.doi.org/10.1038/nrd1384

Mervis J, 2005, Productivity counts — but the definition is key. Science, vol.309(5735): 726–727. http://dx.doi.org/10.1126/science.309.5735.726

David E, Tramontin T and Zemmel R, 2009, Pharmaceutical R&D: the road to positive returns. Nature Reviews Drug Discovery, vol.8(8): 609–610. http://dx.doi.org/10.1038/nrd2948

Cook D, Brown D, Alexander R, et al. 2014, Lessons learned from the fate of AstraZeneca’s drug pipeline: A five-dimensional framework. Nature Reviews Drug Discovery, vol.13: 419–431. http://dx.doi.org/10.1038/nrd4309

DiMasi J A, Hansen R W and Grabowski H G, 2003, The price of innovation: New estimates of drug development costs. Journal of Health Economics, vol.22(2): 151–185. http://dx.doi.org/10.1016/S0167-6296(02)00126-1

DiMasi J A, Feldman L, Seckler A, et al. 2010, Trends in risks associated with new drug development: Success rates for investigational drugs. Clinical Pharmacology and Therapeutics, vol.87(3): 272–277. http://dx.doi.org/10.1038/clpt.2009.295

Basavaraj S and Betageri G V, 2014, Can formulation and drug delivery reduce attrition during drug discovery and development — review of feasibility, benefits and chal-lenges. Acta Pharmaceutica Sinica B, vol.4(1): 3–17. http://dx.doi.org/10.1016/j.apsb.2013.12.003

Bakhtiar R, 2008, Biomarkers in drug discovery and development. Journal of Pharmacology and Toxicological Methods, vol.57(2): 85–91. http://dx.doi.org/10.1016/j.vascn.2007.10.002

Moreno L and Pearson A D, 2013, How can attrition rates be reduced in cancer drug discovery? Expert Opinion in Drug Discovery, vol.8(4): 363–368. http://dx.doi.org/10.1517/17460441.2013.768984

Kola I and Landis J, 2004, Can the pharmaceutical industry reduce attrition rates? Nature Reviews Drug Discovery, vol.3(8): 711–715. http://dx.doi.org/10.1038/nrd1470

Valentin J P, Keisu M and Hammond T G, 2009, Predicting human adverse drug reactions from nonclinical safety studies, in Clinical Trials Handbook, Gad S C (ed), John Wiley & Sons, Inc., Hoboken, NJ, USA, 87–113.

Stummann T C, Beilmann M, Duker G, et al. 2009, Report and recommendations of the workshop of the European Centre for the validation of alternative methods for drug-induced cardiotoxicity. Cardiovascular Toxicology, vol.9(3): 107–125. http://dx.doi.org/10.1007/s12012-009-9045-3

Laverty H G, Benson C, Cartwright E J, et al. 2011, How can we improve our understanding of cardiovascular safety liabilities to develop medicines? British Journal of Pharmacology, vol.163(4): 675–693. http://dx.doi.org/10.1111/j.1476-5381.2011.01255.x

Valentin J P, Kenna J G, Lainée P, et al. 2012, A literature-based analysis of cardiovascular adverse events impacting on drug development. Journal of Pharmacology and Toxicological Methods, vol.66(2): 173. http://dx.doi.org/10.1016/j.vascn.2012.08.050

Tan D S, Thomas G V, Garrett M D, et al. 2009, Biomarker-driven early clinical trials in oncology: A paradigm shift in drug development. Cancer Journal, vol.15(5): 406–420. http://dx.doi.org/10.1097/PPO.0b013e3181bd0445

Berger S I and Iyengar R, 2011, Role of systems pharmacology in understanding drug adverse events. Wiley Interdisciplinary Reviews: Systems Biology and Medicine, vol.3(2): 129–135. http://dx.doi.org/10.1002/wsbm.114

Stark K L, Gross C, Richardson-Jones J, et al. 2007, A novel conditional knockout strategy applied to serotonin receptors, in Conditional Mutagenesis: An Approach to Disease Models, Volume 178: Handbook of Experimental Pharmacology, Feil R and Metzger D (eds), Springer- Verlag Berlin Heidelberg, Berlin, Germany, 347–363. http://dx.doi.org/10.1007/978-3-540-35109-2_14

Bowes J, Rolf M G, Valentin J P, et al. 2006, Pharmacological and pharmaceutical profiling: New trends, in The Process of New Drug Discovery and Development, 2nd edn, Smith C G and O’Donnell J T (eds), Informa Healthcare, New York, 103–134.

Bowes J, Brown A J, Hamon J, et al. 2012, Reducing safety-related drug attrition: The use of in vitro pharmacological profiling. Nature Reviews Drug Discovery, vol.11: 909–922. http://dx.doi.org/10.1038/nrd3845

ICH S7B: The nonclinical evaluation of the potential for delayed ventricular repolarization (QT interval prolongation) by human pharmaceuticals, n.d., viewed April 16, 2016,

ICHS7A: Safety pharmacology studies for human pharmaceuticals, n.d., viewed April 28, 2016,

Assessment of abuse potential of drugs (draft guidance), n.d., viewed April 30, 2016,

Papoian T, Chiu H J, Elayan I, et al. 2015, Secondary pharmacology data to assess potential off-target activity of new drugs: A regulatory perspective. Nature Reviews Drug Discovery, vol.14: 294–296. http://dx.doi.org/10.1038/nrd3845-c1

Cavero I and Guillon J M, 2014, Safety pharmacology assessment of drugs with biased 5-HT2B receptor agonism mediating cardiac valvulopathy. Journal of Pharmac-ological and Toxicological Methods, vol.69(2): 150–161. http://dx.doi.org/10.1016/j.vascn.2013.12.004

Muller P Y and Milton M N, 2012, The determination and interpretation of the therapeutic index in drug devel-opment. Nature Reviews Drug Discovery, vol.11: 751–761. http://dx.doi.org/10.1038/nrd3801

Wu P, Nielsen T E and Clausen M H, 2016, Small-mol-ecule kinase inhibitors: An analysis of FDA-approved drugs. Drug Discovery Today, vol.21(1): 5–10. http://dx.doi.org/10.1016/j.drudis.2015.07.008

Escobar P A, Kemper R A, Tarca J, et al. 2013, Bacterial mutagenicity screening in the pharmaceutical industry. Mutation Research, vol.752(2): 99–118. http://dx.doi.org/10.1016/j.mrrev.2012.12.002

Hastwell P W, Webster T W, Tate M, et al. 2009, Analysis of 75 marketed pharmaceuticals using the GADD45a- GFP ‘GreenScreen HC’ genotoxicity assay. Mutagenesis, vol.24(5): 455–463. http://dx.doi.org/10.1093/mutage/gep029

Hughes C, Rabinowitz A, Tate M, et al. 2012, Development of a high-throughput Gaussia luciferase reporter assay for the activation of the GADD45a gene by mutagens, promutagens, clastogens, and aneugens. Journal of Bio-molecular Screening, vol.17(10): 1302–1315. http://dx.doi.org/10.1177/1087057112453312

Shi J, Bezabhie R and Szkudlinska A, 2010, Further evaluation of a flow cytometric in vitro micronucleus assay in CHO-K1 cells: A reliable platform that detects micronuclei and discriminates apoptotic bodies. Mutagenesis, vol.25(1): 33–40.

http://dx.doi.org/10.1093/mutage/gep040

Tilmant K, Gerets H H J, De Ron P, et al. 2013, The automated micronucleus assay for early assessment of genotoxicity in drug discovery. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, vol.751(1): 1–11. http://dx.doi.org/10.1016/j.mrgentox.2012.10.011

Muster W, Breidenbach A, Fischer H, et al. 2008, Computational toxicology in drug development. Drug Discovery Today, vol.13(7–8): 303–310. http://dx.doi.org/10.1016/j.drudis.2007.12.007

Merlot C, 2010, Computational toxicology — a tool for early safety evaluation. Drug Discovery Today, vol.15(1–2): 6–22. http://dx.doi.org/10.1016/j.drudis.2009.09.010

Valerio L G, 2009, In silico toxicology for the pharma-ceutical sciences. Toxicology and Applied Pharmacology, vol.241(3): 356–370. http://dx.doi.org/10.1016/j.taap.2009.08.022

Stevens J L and Baker T K, 2009, The future of drug safety testing: Expanding the view and narrowing the fo-cus. Drug Discovery Today, vol.14(3–4): 162–167. http://dx.doi.org/10.1016/j.drudis.2008.11.009

Cavero I and Holzgrefe H, 2014, Comprehensive in vitro proarrhythmia assay, a novel in vitro/in silico paradigm to detect ventricular proarrhythmic liability: a visionary 21st century initiative. Expert Opinion Drug Safety, vol.13(6): 745–758. http://dx.doi.org/10.1517/14740338.2014.915311

Sager P T, Gintant G, Turner J R, et al. 2014, Rechanneling the cardiac proarrhythmia safety paradigm: A meeting report from the Cardiac Safety Research Consortium. The American Heart Journal, vol.167(3): 292–300. http://dx.doi.org/10.1016/j.ahj.2013.11.004

Gintant G, Sager P T and Stockbridge N, 2016, Evolution of strategies to improve preclinical cardiac safety testing. Nature Reviews Drug Discovery, vol.15: 457– 471. http://dx.doi.org/10.1038/nrd.2015.34

Rodriguez B, Carusi A, Abi-Gerges N, et al. 2015, Human-based approaches to pharmacology and cardiology: An interdisciplinary and intersectorial workshop. Europace, euv320. http://dx.doi.org/10.1093/europace/euv320

Morissette P, Nishida M, Trepakova E, et al. 2013 The anesthetized guinea pig: An effective early cardiovascular derisking and lead optimization model. Journal of Pharmacological and Toxicological Methods, vol.68(1): 137–149. http://dx.doi.org/10.1016/j.vascn.2013.04.010

Redfern W S, Ewart E, Lainée P, et al. 2013, Functional assessments in repeat-dose toxicity studies: The art of the possible. Toxicology Research, vol.2(4): 209–234. http://dx.doi.org/10.1039/c3tx20093k

Pointon A, Abi-Gerges N, Cross M J, et al. 2013, Phenotypic profiling of structural cardiotoxins in vitro reveals dependency on multiple mechanisms of toxicity. Toxico-logical Sciences, vol.132(2): 317–326. http://dx.doi.org/10.1093/toxsci/kft005

Clements M and Thomas N, 2014, High-throughput multi-parameter profiling of electrophysiological drug effects in human embryonic stem cell derived cardiomyocytes using multi-electrode arrays. Toxicological Sciences, vol.140(2): 445–461. http://dx.doi.org/10.1093/toxsci/kfu084

Reagan W J, 2010, Troponin as a biomarker of cardiac toxicity: past, present, and future. Toxicologic Pathology, vol.38(7): 1134–1137. http://dx.doi.org/10.1177/0192623310382438

Glineur S F, De Ron P, Hanon E, et al. 2016, Paving the route to plasma miR-208a-3p as an acute cardiac injury biomarker: Preclinical rat data supports its use in drug safety assessment. Toxicological Sciences, vol.149(1): 89–97. http://dx.doi.org/10.1093/toxsci/kfv222

Karakikes I, Chaanine A H, Kang S, et al. 2013, Therapeutic cardiac-targeted delivery of miR-1 reverses pres-sure overload-induced cardiac hypertrophy and attenuates pathological remodeling. Journal of the American Heart Association, vol.2(2): e000078. http://dx.doi.org/10.1161/JAHA.113.000078

Lee D S, Chen J H, Lundy D J, et al. 2015, Defined mi-croRNAs induce aspects of maturation in mouse and hu-man embryonic-stem-cell-derived cardiomyocytes. Cell Reports, vol.12(12): 1960–1967. http://dx.doi.org/10.1016/j.celrep.2015.08.042

Kuppusamy K T, Jones D C, Sperber H, et al. 2015, Let-7 family of microRNA is required for maturation and adult-like metabolism in stem cell-derived cardiomyo-cytes. Proceedings of the National Academy of Sciences USA, vol.112(21): E2785–E2794. http://dx.doi.org/10.1073/pnas.1424042112

Olson H, Betton G, Robinson D, et al. 2000, Concordance of the toxicity of pharmaceuticals in humans and in ani-mals. Regulatory Toxicology and Pharmacology, vol.32(1): 56–67. http://dx.doi.org/10.1006/rtph.2000.1399

Greaves P, Williams A and Eve M, 2004, First dose of potential new medicines to humans: How animals help. Nature Reviews Drug Discovery, vol.3: 226–236. http://dx.doi.org/10.1038/nrd1329

Force T and Kerkelä R, 2008, Cardiotoxicity of the new cancer therapeutics — mechanisms of, and approaches to, the problem. Drug Discovery Today, vol.13(17–18): 778–784.

http://dx.doi.org/10.1016/j.drudis.2008.05.011

Patyna S, Arrigoni C, Terron A, et al. 2008, Nonclinical safety evaluation of sunitinib: A potent inhibitor of VEGF, PDGF, KIT, FLT3, and RET receptors. Toxicologic Pa-thology, vol.36(7): 905–916. http://dx.doi.org/10.1177/0192623308326151

Leong X F, Ng C Y and Jaarin K, 2015, Animal models in cardiovascular research: hypertension and atherosclerosis. BioMed Research International, vol.2015(2015): 528757. http://dx.doi.org/10.1155/2015/528757

Authier S, Arezzo J, Delatte M S, et al. 2016, Safety pharmacology investigations on the nervous system: An industry survey. Journal of Pharmacological and Toxicological Methods. http://dx.doi.org/10.1016/j.vascn.2016.06.001

Chikhale E G, Ng K-Y, Burton P S, et al. 1994, Hydrogen bonding potential as a determinant of the in vitro and in situ blood–brain barrier permeability of peptides. Pharmaceutical Research, vol.11(3): 412–419.

http://dx.doi.org/10.1023/A:1018969222130

Deo A, Theil P and Nicolas J M, 2013, Confounding parameters in preclinical assessment of blood-brain-barrier permeation: An overview with emphasis on species differences and effect of disease state. Molecular Pharmaceutics, vol.10(5): 1581–1595. http://dx.doi.org/10.1021/mp300570z

Nicolas J M, 2015, Species differences and impact of disease state on BBB, in Blood-Brain Barrier in Drug Discovery: Optimizing Brain Exposure of CNS Drugs and Minimizing Brain Side Effects for Peripheral Drugs, Di L, Kerns H E (eds), John Wiley & Sons, New Jersey, 66–93. http://dx.doi.org/10.1002/9781118788523.ch4

Easter A, Bell M E, Damewood Jr J R, et al. 2009, Approaches to seizure risk assessment in preclinical drug discovery. Drug Discovery Today, vol.14(17–18): 876– 884. http://dx.doi.org/10.1016/j.drudis.2009.06.003

Bassett L, Troncy E, Pouliot M, et al. 2014, Telemetry video-electroencephalography (EEG) in rats, dogs and non-human primates: Methods in follow-up safety pharmacology seizure liability assessments. Journal of Pharmacological and Toxicological Methods, vol.70(3): 230–240. http://dx.doi.org/10.1016/j.vascn.2014.07.005

Metea M, Litwak M and Arezzo J, 2015, Assessment of seizure risk in pre-clinical studies: Strengths and limitations of the electroencephalogram (EEG). Journal of Pharmacology and Toxicology Methods, vol.75: 135–142. http://dx.doi.org/10.1016/j.vascn.2015.04.001

Muthas D, Boyer S and Hasselgren C, 2013, A critical assessment of modeling safety-related drug attrition. Med-icinal Chemistry Communications, vol.4: 1058–1065. http://dx.doi.org/10.1039/c3md00072a

Arezzo J C, Litwak M S and Zotova E G, 2011, Correlation and dissociation of electrophysiology and histopathology in the assessment of toxic neuropathy. Toxicologic Pathology, vol.39(1): 46–51. http://dx.doi.org/10.1177/0192623310390231

Fix A S and Garman R H, 2000, Practical aspects of neu-ropathology: A technical guide for working with the nervous system. Toxicologic Pathology, vol.28(1): 122– 131. http://dx.doi.org/10.1177/019262330002800115

Garman R H, 2011, Histology of the central nervous system. Toxicologic Pathology, vol.39(1): 22–35. http://dx.doi.org/10.1177/0192623310389621

Kofler J and Wiley C A, 2011, Microglia: key innate immune cells of the brain. Toxicologic Pathology, vol.39(1): 103–114. http://dx.doi.org/10.1177/0192623310387619

Garman R H, 2003, Evaluation of large-sized brains for neurotoxic endpoints. Toxicologic Pathology, vol.31(1): 32–43. http://dx.doi.org/10.1080/01926230390174913

Bolon B, Bradley A, Butt M, et al. 2011, Compilation of international regulatory guidance documents for neuropathology assessment during nonclinical general toxicity and specialized neurotoxicity studies. Toxicologic Pathology, vol.39(1): 92–96. http://dx.doi.org/10.1177/0192623310385145

Switzer III R C, Lowry-Franssen C and Benkovic S A, 2011, Recommended neuroanatomical sampling practices for comprehensive brain evaluation in nonclinical safety studies. Toxicologic Pathology, vol.39(1): 73–84. http://dx.doi.org/10.1177/0192623310397557

Mondello S, Gabrielli A, Catani S, et al. 2012, Increased levels of serum MAP-2 at 6-months correlate with improved outcome in survivors of severe traumatic brain injury. Brain Injury, vol.26(13–14): 1629–1635. http://dx.doi.org/10.3109/02699052.2012.700083

Hanig J, Paule M G, Ramu J, et al. 2014, The use of MRI to assist the section selections for classical pathology as-sessment of neurotoxicity. Regulatory Toxicology and Pharmacology, vol.70(3): 641–647. http://dx.doi.org/10.1016/j.yrtph.2014.09.010

Roberts R A, Aschner M, Calligaro D, et al. 2015, Trans-lational biomarkers of neurotoxicity: A Health and Envi-ronmental Sciences Institute perspective on the way forward. Toxicological Sciences, vol.148(2): 332–340. http://dx.doi.org/10.1093/toxsci/kfv188

Ernst F R and Grizzle A J 2001, Drug-related morbidity and mortality: Updating the cost-of-illness model. Journal of the American Pharmaceutical Association, vol.41(2): 192–199.

Zhou Y, Yang L, Liao Z, et al. 2013, Epidemiology of drug-induced liver injury in China: A systematic analysis of the Chinese literature including 21 789 patients. European Journal of Gastroenterology & Hepatology, vol. 25(7): 825–829. http://dx.doi.org/10.1097/MEG.0b013e32835f6889

Pessayre L D and Biour M, 1999, Drug-induced liver injury, in Oxford Textbook of Clinical Hepatology, 2nd edn, Bircher J, Benhamou J P, McIntyre N, et al. (eds), Oxford, Oxford University Press, 1261–1315.

Park B K, Kitteringham N R, Powell H, et al. 2000, Advances in molecular toxicology towards understanding idiosyncratic drug toxicity. Toxicology, vol.153(1–3): 39–60. http://dx.doi.org/10.1016/S0300-483X(00)00303-6

Roth R A, Luyendyk J P, Maddox J F, et al. 2003, Inflammation and drug idiosyncrasy — is there a connection? The Journal of Pharmacology and Experimental Therapeutics, vol.307(1): 1–8. http://dx.doi.org/10.1124/jpet.102.041624

US Department of Health and Human Services — Food and Drug Administration, n.d., Guidance for industry. Drug-induced liver injury: Premarketing clinical evaluation, viewed February 15, 2016,

Walker R M and McElligott T F, 1981, Furosemide induced hepatotoxicity. Journal of Pathology, vol.135(4): 301–314. http://dx.doi.org/10.1002/path.1711350407

Lee W M, 2003, Drug-induced hepatotoxicity. The New England Journal of Medicine, vol.349(5): 474–485.

http://dx.doi.org/10.1056/NEJMra021844

Godoy P, Hewitt N J, Albrecht U, et al. 2013, Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating me-chanisms of hepatotoxicity, cell signaling and ADME. Archives of Toxicology, vol.87(8): 1315–1530. http://dx.doi.org/10.1007/s00204-013-1078-5

Antoine D J, Williams D P and Park B K, 2008, Under-standing the role of reactive metabolites in drug-induced hepatotoxicity: state of the science. Expert Opinion on Drug Metabolism & Toxicology, vol. 4(11): 1415–1427. http://dx.doi.org/10.1517/17425255.4.11.1415

Usui T, Mise M, Hashizume T, et al. 2009, Evaluation of the potential for drug-induced liver injury based on in vitro covalent binding to human liver proteins. Drug Metabolism and Disposition, vol.37(12): 2383–2392. http://dx.doi.org/10.1124/dmd.109.028860

Obach R S, Kalgutkar A S, Soglia J R, et al. 2008, Can in vitro metabolism-dependent covalent binding data in liver microsomes distinguish hepatotoxic from non-hepatotoxic drugs? An analysis of 18 drugs with consideration of intrinsic clearance and daily dose. Chemical Research in Toxicology, vol.21(9): 1814–1822.

http://dx.doi.org/10.1021/tx800161s

Park B K, Boobis A, Clarke S, et al. 2011, Managing the challenge of chemically reactive metabolites in drug dev-elopment. Nature Reviews Drug Discovery, vol.10(4): 292–306. http://dx.doi.org/10.1038/nrd3408

Williams D P, 2006, Toxicophores: Investigations in drug safety. Toxicology, vol.226(1): 1–11. http://dx.doi.org/10.1016/j.tox.2006.05.041

Ainscow E K, Pilling J E, Brown N M, et al. 2008, Investigations into the liver effects of ximelagatran using high content screening of primary human hepatocyte cultures. Expert Opinion on Drug Safety, vol.7(4): 351–365. http://dx.doi.org/10.1517/14740338.7.4.351

Gerets H H, Tilmant K, Gerin B, et al. 2012, Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biology and Toxicology, vol.28(2): 69–87. http://dx.doi.org/10.1007/s10565-011-9208-4

Persson M, Løye A F, Mow T, et al. 2013, A high content screening assay to predict human drug-induced liver injury during drug discovery. Journal of Pharmacological and Toxicological Methods, vol.68(3): 302–313. http://dx.doi.org/10.1016/j.vascn.2013.08.001

Atienzar F A, Blomme E A, Chen M, et al., 2016, Key challenges and opportunities associated with the use of in vitro models to detect human DILI: Integrated risk assessment and mitigation plans. Accepted June 22, 2016.

Thompson R A, Isin E M, Li Y, et al. 2012, In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs. Chemical Research in Toxicology, vol.25(8): 1616–1632. http://dx.doi.org/10.1021/tx300091x

Chen M, Tung C W, Shi Q, et al. 2014, A testing strategy to predict risk for drug-induced liver injury in humans using high-content screen assays and the ‘rule-of-two’ model. Archives of Toxicology, vol.88(7): 1439–1449. http://dx.doi.org/10.1007/s00204-014-1276-9

Shah F, Leung L, Barton H A, et al. 2015, Setting clinical exposure levels of concern for drug-induced liver injury (DILI) using mechanistic in vitro assays. Toxicological Sciences, vol.147(2): 500–514. http://dx.doi.org/10.1093/toxsci/kfv152

Maschmeyer I, Lorenz A K, Schimek K, et al. 2015, A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents. Lab on a Chip, vol.15(12): 2688–2699. http://dx.doi.org/10.1039/C5LC00392J

Atienzar F A, Novik E I, Gerets H H, et al. 2014, Predictivity of dog co-culture model, primary human hepatocytes and HepG2 cells for the detection of hepatotoxic drugs in humans. Toxicology and Applied Pharmacology, vol.275(1): 44–61. http://dx.doi.org/10.1016/j.taap.2013.11.022

Przybylak K R and Cronin M T, 2012, In silico models for drug-induced liver injury — current status. Expert Opinion in Drug Metabolism and Toxicology, vol.8(2): 201–217. http://dx.doi.org/10.1517/17425255.2012.648613

Zhang M, Chen M and Tong W, 2012, Is toxicogenomics a more reliable and sensitive biomarker than conventional indicators from rats to predict drug-induced liver injury in humans? Chemical Research in Toxicology, vol.25(1): 122–129. http://dx.doi.org/10.1021/tx200320e

Low Y, Uehara T, Minowa Y, et al. 2011, Predicting drug-induced hepatotoxicity using QSAR and toxicogenomics approaches. Chemical Research in Toxicology, vol.24(8): 1251–1262. http://dx.doi.org/10.1021/tx200148a

Regev A, 2014, Drug-induced liver injury and drug de-velopment: Industry perspective. Seminars in Liver Dis-ease, vol.34(2): 227–239. http://dx.doi.org/10.1055/s-0034-1375962

Wang K, Zhang S, Marzolf B, et al. 2009, Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proceedings of the National Academy of Sciences USA, vol.106(11): 4402–4407. http://dx.doi.org/10.1073/pnas.0813371106

Bala S, Petrasek J, Mundkur S, et al. 2012, Circulating microRNAs in exosomes indicate hepatocyte injury and inflammation in alcoholic, drug-induced, and inflammatory liver diseases. Hepatology, vol.56(5): 1946–1957. http://dx.doi.org/10.1002/hep.25873

McGill M R and Jaeschke H, 2014, Mechanistic bio-markers in acetaminophen-induced hepatotoxicity and acute liver failure: From preclinical models to patients. Expert Opinion on Drug Metabolism & Toxicology, vol.10(7): 1005–1017. http://dx.doi.org/10.1517/17425255.2014.920823

Amacher D E, Schomaker S J and Aubrecht J, 2013, Development of blood biomarkers for drug-induced liver injury: An evaluation of their potential for risk assessment and diagnostics. Molecular Diagnosis & Therapy, vol.17(6): 343–354. http://dx.doi.org/10.1007/s40291-013-0049-0

Al-Saffar A, Da Costa A N, Delaunois A, et al. 2015, Gastrointestinal safety pharmacology in drug discovery and development, in Principles of Safety Pharmacology, Handbook of Experimental Pharmacology, Pugsley M K and Curtis M J (eds), Springer Berlin Heidelberg, Berlin, 291–321. http://dx.doi.org/10.1007/978-3-662-46943-9_12

Daniel E E, Kwan C Y and Janssen L, 2001, Pharmacological techniques for the in vitro study of intestinal smooth muscles. Journal of Pharmacological and Toxicological Methods, vol.45(2): 141–158. http://dx.doi.org/10.1016/S1056-8719(01)00131-9

Mahar K M, Portelli S, Coatney R, et al. 2012, Gastric pH and gastric residence time in fasted and fed conscious beagle dogs using the Bravo® pH system. The Journal of Pharmaceutical Sciences, vol.101(7): 2439– 2448. http://dx.doi.org/10.1002/jps.23159

Boillat C S, Gaschen F P, Gaschen L, et al. 2010, Variability associated with repeated measurements of gastrointestinal tract motility in dogs obtained by use of a wireless motility capsule system and scintigraphy. American Journal of Veterinary Research, vol.71(8): 903–908. http://dx.doi.org/10.2460/ajvr.71.8.903

Goineau S, Rompion S, Guillaume P, et al. 2013, Using telemetry to automate the detection of emesis in the ferret: New vistas for delayed emesis assessment. Journal of Pharmacological and Toxicological Methods, vol.68(1): 160–165. http://dx.doi.org/10.1016/j.vascn.2013.03.010

Lesniewska V, Rowland I, Laerke H N, et al. 2006, Relationship between dietary-induced changes in intestinal commensal microflora and duodenojejunal myoelectric activity monitored by radiotelemetry in the rat in vivo. Experimental Physiology, vol.91(1): 229–237. http://dx.doi.org/10.1113/expphysiol.2005.031708

Benjamin A, Da Costa A., Delaunois A, et al. 2015, Renal safety pharmacology in drug discovery and development, in Principles of Safety Pharmacology, Handbook of Experimental Pharmacology, Pugsley M K and Curtis M J(eds), Springer Berlin Heidelberg, Berlin, 323–352. http://dx.doi.org/10.1007/978-3-662-46943-9_13

Dieterle F, Perentes E, Cordier A, et al. 2010, Urinary clusterin, cystatin C, β2-microglobulin and total protein as markers to detect drug-induced kidney injury. Nature Biotechnology, vol.28: 463–469. http://dx.doi.org/10.1038/nbt.1622

Dieterle F, Sistare F, Goodsaid F, et al. 2010, Renal biomarker qualification submission: A dialog between the FDA-EMEA and Predictive Safety Testing Consortium. Nature Biotechnology, vol.28: 455–462. http://dx.doi.org/10.1038/nbt.1625

Vickers A E M, Rose K, Fisher R, et al. 2004, Kidney slices of human and rat to characterize cisplatin-induced injury on cellular pathways and morphology. Toxicologic Pathology, vol.32(5): 577–590. http://dx.doi.org/10.1080/01926230490508821

Husted S, Emanuelsson H, Heptinstall S, et al. 2006, Pharmacodynamics, pharmacokinetics, and safety of the oral reversible P2Y12 antagonist AZD6140 with aspirin in patients with atherosclerosis: A double-blind comparison to clopidogrel with aspirin. European Heart Journal, vol. 27(9): 1038–1047. http://dx.doi.org/10.1093/eurheartj/ehi754

Van Giezen J J, Nilsson L, Berntsson P, et al. 2009, Ticagrelor binds to human P2Y12 independently from ADP but antagonizes ADP-induced receptor signaling and platelet aggregation. Journal of Thrombosis and Haemostasis, vol.7(9): 1556–1565. http://dx.doi.org/10.1111/j.1538-7836.2009.03527.x

Wallentin L, Becker R C, Budaj A, et al. 2009, Ticagrelor versus clopidogrel in patients with acute coronary syndromes. The New England Journal of Medicine, vol.361(11): 1045–1057. http://dx.doi.org/10.1056/NEJMoa0904327

Cannon C P, Husted S, Harrington R A, et al. 2007, Safety, tolerability, and initial efficacy of AZD6140, the first reversible oral adenosine diphosphate receptor antagonist, compared with clopidogrel, in patients with non- ST-segment elevation acute coronary syndrome: Primary results of the DISPERSE-2 trial. Journal of the American College of Cardiology, vol.50(19): 1844–1851.

http://dx.doi.org/10.1016/j.jacc.2007.07.053

Storey R F, Bliden K P, Patil S B, et al. 2010, Incidence of dyspnea and assessment of cardiac and pulmonary function in patients with stable coronary artery disease receiving ticagrelor, clopidogrel, or placebo in the ONSET/ OFFSET study. Journal of the American College of Cardiology, vol.56(3): 185–193. http://dx.doi.org/10.1016/j.jacc.2010.01.062

Nylander S, Femia EA, Scavone M, et al. 2013, Ticagrelor inhibits human platelet aggregation via adenosine in addition to P2Y12 antagonism. Journal of Thrombosis and Haemostasis, vol.11(10): 1867–1876.

http://dx.doi.org/10.1111/jth.12360

Van Giezen J J, Sidaway J, Glaves P, et al. 2012, Ticagrelor inhibits adenosine uptake in vitro and enhances adenosine-mediated hyperemia responses in a canine model. Journal of Cardiovascular Pharmacology and Therapeutics, vol.17(2): 164–172. http://dx.doi.org/10.1177/1074248411410883

Wittfeldt A, Emanuelsson H, Brandrup-Wognsen G, et al. 2013, Ticagrelor enhances adenosine-induced coronary vasodilatory responses in humans. Journal of the American College of Cardiology, vol.61(7): 723–727. http://dx.doi.org/10.1016/j.jacc.2012.11.032

Armstrong D, Summers C, Ewart L, et al. 2014, Charac-terization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. Journal of Cardiovascular Pharmacology and Therapeutics, vol.19(2): 209–219. http://dx.doi.org/10.1177/1074248413511693

Nicolas J M, Chanteux H, Mancel V, et al. 2014, N-alkylprotoporphyrin formation and hepatic porphyria in dogs after administration of a new antiepileptic drug candidate: Mechanism and species specificity. Toxicological Sciences, vol.141(2): 353–364. http://dx.doi.org/10.1093/toxsci/kfu131

Balwani M and Desnick R J, 2012, The porphyrias: Advances in diagnosis and treatment. Blood, vol.120(23): 4496–4504. http://dx.doi.org/10.1182/blood-2012-05-423186

Lavigne J A, Nakatsu K and Marks G S, 2002, Identification of human hepatic cytochrome P450 sources of N-alkylprotoporphyrin IX after interaction with porphy-rinogenic xenobiotics, implications for detection of xenobiotic-induced porphyria in humans. Drug Metabolism and Disposition, vol.30(7): 788–794.

http://dx.doi.org/10.1124/dmd.30.7.788

McNamee J P, Jurima-Romet M, Kobus S M, et al. 1997, cDNA-expressed human cytochrome P450 isozymes. Inactivation by porphyrinogenic xenobiotics. Drug Metabolism and Disposition, vol.25(4): 437–441.

Bailey J, Thew M and Balls M, 2015, Predicting human drug toxicity and safety via animal tests: Can any one species predict drug toxicity in any other, and do monkeys help? Alternatives to Laboratory Animals, vol.43(6): 393–403.

Ewart L, Aylott M, Deurinck M, et al. 2014, The concordance between nonclinical and phase I clinical cardiovascular assessment from a cross-company data sharing initiative. Toxicological Sciences, vol.142(2): 427–435. http://dx.doi.org/10.1093/toxsci/kfu198

Leist M and Hartung T, 2013, Inflammatory findings on species extrapolations: Humans are definitely no 70-kg mice. Archives of Toxicology, vol.87(4): 563–567. http://dx.doi.org/10.1007%2Fs00204-013-1038-0

Khatri P, Sirota M and Butte A J, 2012, Ten years of pathway analysis: Current approaches and outstanding challenges. PLoS Computational Biology, vol.8: e1002375. http://dx.doi.org/10.1371/journal.pcbi.1002375

Krejsa C M, Horvath D, Rogalski S L, et al. 2003, Predicting ADME properties and side effects: The BioPrint approach. Current Opinion in Drug Discovery Development, vol.6: 470–480.

Fliri A F, Loging W T, Thadeio P F, et al. 2005, Biological spectra analysis: Linking biological activity profiles to molecular structure. Proceedings of the National Academy of Sciences of the USA, vol.102: 261–266. http://dx.doi.org/10.1073/pnas.0407790101

Bender A, Scheiber J, Glick M, et al. 2007, Analysis of pharmacology data and the prediction of adverse drug reactions and off-target effects from chemical structure. ChemMedChem, vol.2(6): 861–873. http://dx.doi.org/10.1002/cmdc.200700026

Keiser M J, Roth B L, Armbruster B N, et al. 2007, Re-lating protein pharmacology by ligand chemistry. Nature Biotechnology, vol.25: 197–206. http://dx.doi.org/10.1038/nbt1284

Garcia-Serna R and Mestres J, 2010, Anticipating drug side effects by comparative pharmacology. Expert Opinion on Drug Metabolism and Toxicology, vol.6(10): 1253–1263. http://dx.doi.org/10.1517/17425255.2010.509343

Garcia-Serna R, Vidal D, Remez N, et al. 2015, Large-scale predictive drug safety: From structural alerts to biological mechanisms. Chemical Research in Toxicology, vol.28(10): 1875–1887. http://dx.doi.org/10.1021/acs.chemrestox.5b00260

Wallace K B and Starkov A A, 2000, Mitochondrial targets of drug toxicity. Annual Review of Pharmacology and Toxicology, vol.40: 353–388. http://dx.doi.org/10.1146/annurev.pharmtox.40.1.353

Wallace K B, 2008, Mitochondrial off targets of drug therapy. Trends in Pharmacological Sciences, vol.29(7): 361–366. http://dx.doi.org/10.1016/j.tips.2008.04.001

Pereira C V, Moreira A C, Pereira S P, et al. 2009, Investigating drug-induced mitochondrial toxicity: A biosensor to increase drug safety? Current Drug Safety, vol.4(1): 34–54. http://www.eurekaselect.com/68326/article#

Labbe G, Pessayre D and Fromenty B, 2008, Drug-induced liver injury through mitochondrial dysfunction: Mechanisms and detection during preclinical safety studies. Fundamental & Clinical Pharmacology, vol.22(4): 335–353. http://dx.doi.org/10.1111/j.1472-8206.2008.00608.x

Porceddu M, Buron N, Roussel C, et al. 2012, Prediction of liver injury induced by chemicals in human with a multiparametric assay on isolated mouse liver mitochondria. Toxicological Sciences, vol.129(2): 332–345.

http://dx.doi.org/10.1093/toxsci/KFS197

Wallace K B, 2003, Doxorubicin-induced cardiac mitochondrionopathy. Pharmacology & Toxicology, vol.93(3): 105–115. http://dx.doi.org/10.1034/j.1600-0773.2003.930301.x

Finsterer J and Ohnsorge P, 2013, Influence of mitochondrion-toxic agents on the cardiovascular system. Regulatory Toxicology and Pharmacology, vol.67(3): 434–445. http://dx.doi.org/10.1016/j.yrtph.2013.09.002

Beeson C C, Beeson G C and Schnellmann R G, 2010, A high-throughput respirometric assay for mitochondrial biogenesis and toxicity. Analytical Biochemistry, vol.404(1): 75–81. http://dx.doi.org/10.1016/j.ab.2010.04.040

Dykens J A and Will Y (eds), 2008, Drug-induced Mitochondrial Dysfunction. New Jersey: John Wiley and Sons, Inc. http://dx.doi.org/10.1002/9780470372531

Honkoop P, Scholte H R, De Man R A, et al. 1997, Mitochondrial injury: Lessons from the fialuridine trial. Drug Safety, vol.17(1): 1–7. http://dx.doi.org/10.2165/00002018-199717010-00001

Orlowski J P, 1999, Whatever happened to Reye’s syndrome? Did it ever really exist? Critical Care Medicine, vol.27(8): 1582–1587. http://dx.doi.org/10.1097/00003246-199908000-00032

Zhang J, Nuebel E, Wisidagama D R, et al. 2012, Measuring energy metabolism in cultured cells, including human pluripotent stem cells and differentiated cells. Nature Protocols, vol.7(6): 1068–1085. http://dx.doi.org/10.1038/nprot.2012.048

Nelms M D, Mellor C L, Cronin M T, et al. 2015, Development of an in silico profiler for mitochondrial toxicity. Chemical Research in Toxicology, vol.28(10): 1891–1902.

http://dx.doi.org/10.1021/acs.chemrestox.5b00275

Benard G and Rossignol R, 2008, Ultrastructure of the mitochondrion and its bearing on function and bioenergetics. Antioxidants & Redox Signaling, vol.10(8): 1313–1342. http://dx.doi.org/10.1089/ars.2007.2000

Ferrick D A, Neilson A and Beeson C, 2008, Advances in measuring cellular bioenergetics using extracellular flux. Drug Discovery Today, vol.13(5–6): 268–274. http://dx.doi.org/10.1016/j.drudis.2007.12.008

Wills L P, Beeson G C, Trager R E, et al. 2013, High-throughput respirometric assay identifies predictive toxicophore of mitochondrial injury. Toxicology and Applied Pharmacology, vol.272(2): 490–502. http://dx.doi.org/10.1016/j.taap.2013.06.014

Nicholls D G, Darley-Usmar V M, Wu M, et al. 2010, Bioenergetic profile experiment using C2C12 myoblast cells. The Journal of Visualized Experiments, vol.46: e2511. http://dx.doi.org/10.3791/2511

Kola I, 2008, The state of innovation in drug development. Clinical Pharmacology and Therapeutics, vol.83(2): 227–230. http://dx.doi.org/10.1038/sj.clpt.6100479

Leurent C and Ehlers M D, 2015, Digital technologies for cognitive assessment to accelerate drug development in Alzheimer's disease. Clinical Pharmacology & Therapeutics, vol.98(5): 475–476. http://dx.doi.org/10.1002/cpt.212

Cobice D F, Goodwin R J A, Andren P E, et al. 2015, Future technology insight: Mass spectrometry imaging as a tool in drug research and development. British Journal of Pharmacology, vol.172(13): 3266–3283. http://dx.doi.org/10.1111/bph.13135

Atienzar F A, Gerets H, Tilmant K, et al. 2013, Evaluation of impedance-based label-free technology as a tool for pharmacology and toxicology investigations. Biosensors, vol.3(1): 132–156. http://dx.doi.org/10.3390/bios3010132

Atienzar F A, Tilmant K, Gerets H H, et al. 2011, The use of real-time cell analyzer technology in drug discovery: Defining optimal cell culture conditions and assay reproducibility with different adherent cellular models. Journal of Biomolecular Screening, vol.16(6): 575–587. http://dx.doi.org/10.1177/1087057111402825

Abassi Y A, Xi B, Zhang W, et al. 2009, Kinetic cell-based morphological screening: Prediction of mechanism of compound action and off-target effects. Chemistry and Biology, vol.16(7): 712–723. http://dx.doi.org/10.1016/j.chembiol.2009.05.011

Kirstein S L, Atienza J M, Xi B, et al. 2006, Live cell quality control and utility of real-time cell electronic sensing for assay development. Assay and Drug Development Technologies, vol.4(5): 545–553. http://dx.doi.org/10.1089/adt.2006.4.545

Caplin J D, Granados N G, James M R, et al. 2015, Microfluidic organ-on-a-chip technology for advancement of drug development and toxicology. Advanced Healthcare Materials, vol.4(10): 1426–1450. http://dx.doi.org/10.1002/adhm.201500040

Stucki A O, Stucki J D, Hall S R R, et al. 2015, A lung-on-a-chip array with an integrated bio-inspired respiration mechanism. Lab on a Chip, vol.15(5): 1302–1310. http://dx.doi.org/10.1039/C4LC01252F

Khetani S R and Bhatia S N, 2008, Microscale culture of human liver cells for drug development. Nature Biotech-nology, vol.26(1): 120–126. http://dx.doi.org/10.1038/nbt1361

Khetani S R, Kanchagar C, Ukairo O, et al. 2013, Use of micropatterned cocultures to detect compounds that cause drug-induced liver injury in humans. Toxicological Sciences, vol.132(1): 107–117. http://dx.doi.org/10.1093/toxsci/kfs326

Ukairo O, Kanchagar C, Moore A, et al. 2013, Long-term stability of primary rat hepatocytes in micropatterned cocultures. Journal of Biochemical and Molecular Toxicology, vol.27(3): 204–212. http://dx.doi.org/10.1002/jbt.21469

Itzhaki I, Maizels L, Huber I, et al. 2011, Modelling the long QT syndrome with induced pluripotent stem cells. Nature, vol.471: 225–229. http://dx.doi.org/10.1038/nature09747

Sun N, Yazawa M, Liu J, et al. 2012, Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Science Translational Medicine, vol.4(130): 130–147. http://dx.doi.org/10.1126/scitranslmed.3003552

Lan F, Lee A S, Liang P, et al. 2013, Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell, vol.12(1): 101–113. http://dx.doi.org/10.1016/j.stem.2012.10.010

Saumarez R C, Chojnowska L, Derksen R, et al. 2003, Sudden death in noncoronary heart disease is associated with delayed paced ventricular activation. Circulation, vol.107: 2595–2600. http://dx.doi.org/10.1161/01.cir.0000068342.96569.a1

Liang P, Lan F, Lee AS, et al. 2013, Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation, vol.127(16): 1677–1691. http://dx.doi.org/10.1161/CIRCULATIONAHA.113.001883

Brennan F R, Morton L D, Spindeldreher S, et al. 2010, Safety and immunotoxicity assessment of immunomodulatory monoclonal antibodies. Monoclonal Antibodies, vol.2(3): 233–255. http://dx.doi.org/10.4161/mabs.2.3.11782

Muller P Y, Milton M, Lloyd P, et al. 2009, The minimum anticipated biological effect level (MABEL) for selection of first human dose in clinical trials with monoclonal an-tibodies. Current Opinion in Biotechnology, vol.20(6): 722–729. http://dx.doi.org/10.1016/j.copbio.2009.10.013

Heyen J R, Rojko J, Evans M, et al. 2014, Characterization, biomarkers, and reversibility of a monoclonal antibody-induced immune complex disease in cynomolgus monkeys (Macaca fascicularis). Toxicologic Pathology, vol.42(4): 765–773. http://dx.doi.org/10.1177/0192623314522559

Hillebrecht A, Muster W, Brigo A, et al. 2011, Comparative evaluation of in silico systems for Ames test mutagenicity prediction: Scope and limitations. Chemical Research in Toxicology, vol.24(6): 843–854. http://dx.doi.org/10.1021/tx2000398

Guo L, Coyle L, Abrams R M, et al. 2013, Refining the human iPSC-cardiomyocyte arrhythmic risk assessment model. Toxicological Sciences, vol.136(2): 581–594. http://dx.doi.org/10.1093/toxsci/kft205

Kirkland D, Aardema M, Henderson L, et al. 2005, Evaluation of the ability of a battery of three in vitro genotoxicity tests to discriminate rodent carcinogens and non-carcinogens I. Sensitivity, specificity and relative predictivity. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, vol.584(1–2): 1–256.

http://dx.doi.org/10.1016/j.mrgentox.2005.02.004

Winter M J, Redfern W S, Hayfield A J, et al. 2008, Validation of a larval zebrafish locomotor assay for assessing the seizure liability of early-stage development drugs. Journal of Pharmacological and Toxicological Methods, vol.57(3): 176–187.

http://dx.doi.org/10.1016/j.vascn.2008.01.004

Mulliner D, Schmidt F, Stolte M, et al. 2016, Computational models for human and animal hepatotoxicity with a global application scope. Chemical Research in Toxicology, vol.29(5): 757–767. http://dx.doi.org/10.1021/acs.chemrestox.5b00465

Beattie K A, Luscombe C, Williams G, et al. 2013, Eval-uation of an in silico cardiac safety assay: Using ion channel screening data to predict QT interval changes in the rabbit ventricular wedge. Journal of Pharmacological and Toxicological Methods, vol.68(1): 88–96. http://dx.doi.org/10.1016/j.vascn.2013.04.004

Wallis R M, 2010, Integrated risk assessment and predictive value to humans of non-clinical repolarization assays. British Journal of Pharmacology, vol.159(1): 115–121. http://dx.doi.org/10.1111/j.1476-5381.2009.00395.x

Harmer A R, Abi-Gerges N, Morton M J, et al. 2012, Validation of an in vitro contractility assay using canine ventricular myocytes. Toxicology and Applied Pharma-cology, vol.260(2): 162–172. http://dx.doi.org/10.1016/j.taap.2012.02.007

Keating C, Martinez V, Ewart L, et al. 2010, The validation of an in vitro colonic motility assay as a biomarker for gastrointestinal adverse drug reactions. Toxicology and Applied Pharmacology, vol.245(3): 299–309. http://dx.doi.org/10.1016/j.taap.2010.03.014

Hornberg J J, Laursen M, Brenden N, et al. 2014, Exploratory toxicology as an integrated part of drug discovery. Part II: Screening strategies. Drug Discovery Today, vol.19(8): 1137–1144. http://dx.doi.org/10.1016/j.drudis.2013.12.009

Jennings P, 2015, The future of in vitro toxicology. Toxicology in Vitro, vol.29(6): 1217–1221. http://dx.doi.org/10.1016/j.tiv.2014.08.011

Downloads

Published

2023-07-25