Introduction
Early in 2018, the UK’s BioIndustry Association and the Medicines Discovery Catapult 1 published a remarkable report calling for the process of drug discovery to be ‘humanised’ in order to ease the productivity crisis in pharmaceutical research. The mission of the Medicines Discovery Catapult is to help UK biotech companies transform so that the UK can maintain its position as one of the prime locations for developing new medicines. The UK industry leaders interviewed for the report stated that the key problem with the current system was the poor ability of existing pre-clinical models to reliably predict safety and efficacy in humans. Human drug-induced toxicities that are poorly predicted by animal safety studies (i.e. animal/human concordance < 60%) include toxicities of the liver,2–4 the heart,5,6 the immune system,2,3 the skin,2,3 the endocrine system2,3 and the central nervous system. 7 Poor human predictivity results in only a small proportion of drug candidates translating into treatments for humans despite having first appeared safe and effective in animal studies, with recent approval rates ranging from 13.8% 8 to 9.6%. 9 To remedy this situation, the BioIndustry Association and Medicines Discovery Catapult report identified an urgent need to ‘retool’ drug discovery with the many human-relevant technologies that have been developed, in order to make the early stages of research more predictive of how drugs work in humans and consequently to have better drug candidates entering human trials, leading to lower rates of attrition.
The UK government’s innovation agency has also recognised the potential of human-relevant approaches for improving drug safety and efficacy. 10 Like the Medicines Discovery Catapult, it identifies pre-clinical animal studies as the problem, observes that companies and regulators are increasingly recognising the limitations of such studies and suggests that new technologies could ultimately replace animals for testing drug efficacy and safety. The agency envisions a thriving ‘non-animal technology’ sector in the UK, with huge market potential and opportunity for driving UK economic growth. However, while noting that the UK has world-leading research in this area as well as companies able to take advantage of new commercial opportunities, it highlights that significant investment in non-animal technologies is taking place in the US and Europe. Indeed the Netherlands, in a bold move, has declared its aim to lead the world in phasing out the use of animals in the regulatory safety testing of medicines and chemicals by a target date of 2025, regarding new technologies as having huge potential to increase research relevance and to deliver more reliable risk assessments, while maintaining existing safety levels.11,12
Human-relevant approaches
The new approaches creating such optimism do not consist of one-by-one replacements of specific animal tests 13 but represent a completely new, human-focused and systems-biology 14 based approach to drug discovery, incorporating a range of in vitro, in silico and human in vivo methodologies. They are driven by the need to reduce costs and increase the speed and accuracy of drug discovery, as well as by an enormous appetite for change. 15 The generation of human-induced pluripotent stem cells (iPSC) has been a significant development; as they possess the genetic background of the individual they can be used to create disease- or patient-specific models, such as ‘organoids’. These are simplified in vitro versions of organs, capable of modelling some specific function of that organ. 16 In the field of cystic fibrosis for example, patient-derived intestinal organoids have been used to select an effective therapy, which was then used to successfully treat the patient. 17 Human-induced pluripotent stem cells are anticipated to transform biological research and healthcare, 18 with stem cell banks being established worldwide and the global market for human-induced pluripotent stem cells technology estimated to reach $2.9 billion in 2018. 10
The creation of micro-physiological systems, known as ‘organs-on-chips’, is another revolutionary development. Organs-on-chips are bio-engineered devices that mimic key aspects of the physiology and function of human organs, replicating some of the complexity of the human body environment on a microscopic scale. They are able to mimic blood, air and nutrient flow, as well as mechanical forces such as peristalsis and can be continuously monitored to obtain a profile over time. Organs-on-chips enable the study of basic biological processes, the modelling of diseases and investigation of the effects of drugs. They can potentially identify safety and efficacy issues earlier and more reliably in the drug development process, enabling the design and selection of drug candidates that are more likely to succeed in human clinical trials. The US is making significant investments in organ-on-chip technologies. 19
The US government is also investing in other new technologies for drug safety screening. For its ‘Tox21’ initiative 10,000 chemicals are being tested using a panel of automated high-throughput human cell-based screening assays. Incorporating data from these assays into computational models can result in models that greatly outperform the predictive ability of models built with animal toxicity data. 20 The global market for cell-based assays in drug discovery, safety and toxicology is expected to reach US$21 billion in 2018. 10 Toxicology is also adopting a new conceptual framework for risk assessment known as Adverse Outcome Pathways. These describe in detail the complex chain of biological processes that occur from the moment a chemical enters the body and enable a more comprehensive understanding of how toxicity is expressed. This new systems biology approach harnesses the power of mathematical modelling to make predictions, for example physiologically based pharmacokinetic mathematical models are used to predict the absorption, distribution, metabolism and excretion of substances. Additionally, human-focused models (such as the cystic fibrosis organoids mentioned above) allow ‘surrogate’ trials to be conducted before advancing to actual clinical trials, providing a vital, physiologically relevant bridge between pre-clinical investigations and clinical outcomes and bringing the possibility of personalised medicine closer.
Emerging evidence of effectiveness
Many of the new technologies, such as human-induced pluripotent stem cells and cell-based assays, are gaining increasingly large scale investment because of the potential they have shown to date; nevertheless they need to be effective, reliable and evidence-based. 13 The intergovernmental Organisation for Economic Cooperation and Development is currently developing guidance on improving the reliability of human in vitro methods. 21 Evidence is only just beginning to emerge but initial comparisons with animal data suggest they may be better at predicting adverse drug reactions. A micro-liver comprising human liver cells, for example, is able to predict liver damage from Fialuridine, the drug that killed five patients in a 1993 clinical trial 22 and many other liver-toxic drugs whose adverse effects had not been predicted by animal tests. 23 A blood vessel chip is able to accurately model and predict thrombosis induced by monoclonal antibody drugs that caused thrombosis in clinical trials, an adverse reaction that had not been predicted in pre-clinical testing. 24 Meanwhile, an innovative in silico ‘drug trial’ tested 62 drugs and reference compounds in more than 1000 simulations of human cardiac cells. The computer model predicted the risk of human drug-induced heart arrhythmias with 89% accuracy, compared with animal studies that showed up to 75% accuracy. 25 Cardiac toxicity can also be predicted in vitro: an assay using primary human heart cells to assess the potential of drugs to disrupt heart rhythm or contractility (two serious liabilities responsible for many drug failures) demonstrated excellent prediction of real clinical outcomes, with 96% sensitivity and 100% specificity in the reference drugs tested. Furthermore, a comparison between human and dog heart cells for two of the test drugs highlighted the inability of canine models (default models for drug cardiac safety assessment) or canine tissues to accurately predict the risk of these adverse effects on the human heart. 26 Table 1 provides further examples of human-relevant methods that can predict human drug toxicities that are poorly predicted by animal studies.
Table 1.
Organ system affected | Human relevant model a | Reference |
---|---|---|
Hepatic | In vitro hazard matrix (cell cytotoxicity + bile salt efflux pump inhibition + mitochondrial toxicity + covalent binding to hepatocyte proteins) | 27,28 |
Micropatterned hepatocyte co-cultures | 29 | |
Human liver spheroids | 30 | |
Hepatocyte high content cell imaging | 31 | |
Quantitative systems toxicology modelling | 32 | |
Cardiac | CiPA panel assays | 6,33 |
CNS | Human-induced pluripotent stem cell-derived neuronal cell cultures | 34 |
The performance of these assays was assessed by testing drugs whose toxicity/lack of toxicity in humans was known.
Emerging evidence also suggests that the new technologies may be more capable of elucidating mechanisms of toxicity than animal studies. For example Bavli et al. 35 used liver-on-chip technology to investigate why Rezulin (a type-2 diabetes drug) had caused unexplained liver damage in clinical trials. Using the chip they found that even low concentrations of Rezulin caused liver stress before any damage was visible. The same team was able to identify a mechanism of toxicity of Paracetamol using similar technology. 36 Van Esbroeck et al. 37 used human cells to investigate the off-target effects of BIA 10-2474, a drug that killed a healthy volunteer and hospitalised four others in France. They found that BIA 10-2474 deactivated multiple proteins and that this caused disruption to the metabolism of human nerve cells, effects that had not been predicted by safety testing in animals.
The evidence so far is very encouraging; however, systematic comparisons of animal versus human-relevant methods would provide more definitive evidence. As a step in this direction, the Evidence Based Toxicology Collaboration (www.ebtox.org) is using systematic review methodology to compare animal and human in vitro data in terms of how well each predicts human adverse effects (on the liver), taking two anti-diabetic drugs (Troglitazone and Rosiglitazone) as case studies. 38 Further evidence will accumulate once there is wider uptake of these technologies. Systematic reviews will be vital in synthesising this evidence to provide high quality comparisons; indeed they are already being used to explore the evidence base for human-relevant technologies in toxicology as the focus shifts from animal to human biology. 39
Ensuring safety during the transition to human-relevant methods
Human safety is the paramount consideration when adopting innovative approaches. A wealth of legacy data on drug safety and test performance is currently being systematically reviewed 40 and as a safeguard during the transition to human-relevant methodologies, this evidence can be used alongside techniques such as ‘read-across’ (using information from a data-rich substance for a data-poor substance that is considered sufficiently similar) and computer modelling 41 to provide a ‘weight of evidence’ approach. This integrated approach to testing and assessment 42 has been developed and accepted as the best method by regulatory agencies in the areas of skin and eye irritation, driven by the ban on animal testing for cosmetics. It has recently been demonstrated that machine-learning software combined with big data can now be used to create sophisticated read-across-based tools that greatly outperform animal studies in predicting chemical safety, with an accuracy of 80%–95%, compared to 50%–70% for the respective animal tests. 43
As a further safeguard, low-risk approaches such as microdosing (whereby drugs are administered in doses small enough to be safe, but large enough for the cellular response to be studied) can be used to aid prediction of drug kinetics and hence drug dose when advancing to first testing in humans. 44
Microdosing is used in a number of ways, including in ‘Phase 0’ trials, in identifying drug-drug interactions and – increasingly – to assess absolute bioavailability (i.e. to determine what fraction of the therapeutic dose actually reaches systemic circulation), helping to establish a safe dosing level. It can also be used to determine the pharmacodynamic activity of a drug, and allows the measurement of drug concentrations in single cells. 45
Clinical trials also need to be longer, larger and more diverse in order to pick up rare adverse drug reactions (which animal studies cannot predict 46 ), and adverse drug reactions and deaths need to be properly reported.47,48 Human-relevant technologies can address some of these issues through the use of human-derived biomarkers to select trial participants.49,50
Validation
New technologies need to be fit-for-purpose. 15 In other words, if they are intended to be used to test drug safety they need to be acceptable to regulators. Unfortunately, however, regulators currently only accept safety data derived from new technologies if the latter have been validated against historical animal data. This makes little sense as the tests must predict how the drugs will behave in humans, not animals, and means that the new tests cannot succeed if the drug in question affects animals differently from humans, which is often the case.13,51,52 An expert group convened by the UK’s National Centre for the Replacement, Refinement and Reduction of Animals in Research recommends that regulatory authorities develop ‘Performance-Based Standards’ aimed directly at human safety rather than at reproducing the results of animal studies. The group also advocates using ‘safe-haven’ trials, where traditional and new methodology data are submitted in parallel to develop experience and regulatory confidence in new methods. 53 Two roadmaps issued by the United States government suggest that validation needs to be more flexible if promising new technologies are to be integrated into the system.54,55 One suggests that some new technologies might be ‘qualified’ for specific and clearly defined contexts of use, exempting them from formal validation procedures. 54 Indeed the US Food and Drug Administration has recently committed to a leading role in qualifying Emulate’s ‘organs-on-chips’ technology for use in regulatory toxicology testing. 19 It has also been suggested that innovators need to start by finding out what regulators need and want, rather than developing a technology and then hoping that regulators can find a use for it. 56 Both US roadmaps emphasise a need for innovators and regulators to communicate to a much greater extent about each other’s needs.54,55
Conclusion
There is clearly great optimism about human-relevant approaches to drug discovery, particularly regarding their potentially greater ability to predict human adverse drug reactions (when compared with animal studies) and their potential to weed out failures earlier on in the drug discovery process. Nevertheless, there are obstacles to reducing our existing reliance on animals. The current research paradigm perpetuates animal-based approaches through ‘lock-in’ mechanisms such as investments in infrastructure, or funding mechanisms. 57 These create ‘path dependence’ and can make change difficult. 58 Similarly regulations can stifle innovation and perpetuate animal-based approaches. Funding incentives could disrupt the current institutional lock-in to animal research by prioritising funding for human-based biomedical research over funding for ‘improved’ animal models. 19 After all, even ‘improved’ animal models introduce uncertainty into the system due to species differences. 59 The funding of systematic reviews in this area would allow high quality evidence to be generated and explored. Regulations could be amended to allow innovation to flourish, as happened in Japan with the development of human-induced pluripotent stem cells technology. 60 Right now there is a tremendous opportunity for the UK to seize the initiative on humanising drug discovery and to revolutionise medicine through more intelligent, human-relevant research. Human-relevant approaches have potential to offer insight into the functioning of the integrated human system, to speed up drug discovery, save money and improve the safety and efficacy of medicines. A shift in focus by funding and regulatory bodies would allow their potential to be fully explored.
Declarations
Competing Interests
KA, GK and PP work for the Safer Medicines Trust which is an independent, charitable patient safety organisation that promotes the use of scientifically valid human-relevant methods and technologies for the development of safe and effective medicines.
Funding
None declared.
Ethics approval
Not applicable.
Guarantor
KA.
Contributorship
KA conceived the idea for this paper, wrote the first draft and contributed to the subsequent drafts before approving the final version. Both KT and GK contributed to the content of the paper and helped redraft it before approving the final version. PP contributed to the conception and design of the paper and produced subsequent drafts for final approval.
Acknowledgements
We are very grateful to the reviewer for his comments and suggestions.
Provenance
Not commissioned; peer-reviewed by Robin Ferner.
References
- 1.BioIndustry Association and Medicines Discovery Catapult. State of the Discovery Nation 2018 and the role of the Medicines Discovery Catapult, https://md.catapult.org.uk/FlipBuilder/mobile/index.html (2018, accessed 26 April 2018).
- 2.Olson H, Betton G, Robinson D, Thomas K, Monro A, Kolaja G, et al. Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol 2000; 32: 56–67. [DOI] [PubMed] [Google Scholar]
- 3.Greaves P, Williams A, Eve M. First dose of potential new medicines to humans: how animals help. Nat Rev Drug Discov 2004; 3: 226–236. [DOI] [PubMed] [Google Scholar]
- 4.Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat Rev Drug Discov 2015; 14: 475–475. [DOI] [PubMed] [Google Scholar]
- 5.Laverty HG, Benson C, Cartwright EJ, Cross MJ, Garland C, Hammond T, et al. How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines? British journal of pharmacology 2011; 163: 675–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gintant G, Sager PT, Stockbridge N. Evolution of strategies to improve preclinical cardiac safety testing. Nat Rev Drug Discov 2016; 15: 457–457. [DOI] [PubMed] [Google Scholar]
- 7.Mead AN, Amouzadeh HR, Chapman K, Ewart L, Giarola A, Jackson SJ, et al. Assessing the predictive value of the rodent neurofunctional assessment for commonly reported adverse events in phase I clinical trials. Regul Toxicol Pharmacol 2016; 80: 348–357. [DOI] [PubMed] [Google Scholar]
- 8.Wong CH, Siah KW, Lo AW. Estimation of clinical trial success rates and related parameters. Biostatistics 2018, pp. 1–14. 10.1093/biostatistics/kxx069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Thomas DW, Burns J, Audette J, Carroll A, Dow-Hygelund C, Hay M. Clinical development success rates 2006–2015, San Diego: Biomedtracker/Washington, DC: BIO/Bend: Amplion, 2016. [Google Scholar]
- 10.Innovate UK. A non-animal technologies roadmap for the UK: advancing predictive biology, https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/474558/Roadmap_NonAnimalTech_final_09Nov2015.pdf (2015, accessed 26 April 2018).
- 11.Netherlands National Committee for the protection of animals used for scientific purposes. Transition to non-animal research: on opportunities for the phasing out of animal procedures and the stimulation of innovation without laboratory animals, www.ncadierproevenbeleid.nl/documenten/rapport/2016/12/15/ncad-opinion-transition-to-non-animal-research (2016, accessed 26 April 2018).
- 12.National Institute for Public Health and the Environment, RIVM. Roadmap for animal-free innovations in regulatory safety assessment, www.rivm.nl/dsresource?objectid=571f1221-8ada-4488-99ef-2424acd50d30 (2018, accessed 26 June 2018).
- 13.Hartung T. Evidence-based toxicology – the toolbox of validation for the 21st century? ALTEX 2010; 27: 253–263. [DOI] [PubMed] [Google Scholar]
- 14.Gan W. Interview with a thought leader on systems medicine—Weiniu Gan, PhD. Syst Med 2018; 1: 9–10. [Google Scholar]
- 15.Burden N, Mahony C, Müller BP, Terry C, Westmoreland C, Kimber I. Aligning the 3Rs with new paradigms in the safety assessment of chemicals. Toxicology 2015; 330: 62–66. [DOI] [PubMed] [Google Scholar]
- 16.Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 2014; 345: 12471251–12471259. [DOI] [PubMed] [Google Scholar]
- 17.McCarthy C, Brewington JJ, Harkness B, Clancy JP, Trapnell BC. Personalised CFTR pharmacotherapeutic response testing and therapy of cystic fibrosis. Eur Respir J 2018; 51: 1702457–1702457. [DOI] [PubMed] [Google Scholar]
- 18.Ronaldson-Bouchard K, Vunjak-Novakovic G. Organs-on-a-chip: a fast track for engineered human tissues in drug development. Cell Stem Cell 2018; 22: 310–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Marshall LJ, Austin CP, Casey W, Fitzpatrick SC, Willett C. Recommendations toward a human pathway-based approach to disease research. Drug Discov Today 2018, pp. 30473–30477. [DOI] [PubMed] [Google Scholar]
- 20.Huang R, Xia M, Sakamuru S, Zhao J, Lynch C, Zhao T, et al. Expanding biological space coverage enhances the prediction of drug adverse effects in human using in vitro activity profiles. Sci Rep 2018; 8: 3783–3783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.OECD. Draft guidance document on good in vitro method practices (GIVIMP) for the development and implementation of in vitro methods for regulatory use in human safety assessment, www.oecd.org/env/ehs/testing/OECD%20Final%20Draft%20GIVIMP.pdf (2017, accessed 11 May 18).
- 22.Baker M. Tissue models: a living system on a chip. Nature 2011; 471: 661–665. [DOI] [PubMed] [Google Scholar]
- 23.Xu J, Henstock P, Dunn M, Smith A, Chabot J, de Graaf D. Cellular imaging predictions of clinical drug-induced liver injury. Toxicol Sci 2008; 105: 97–105. [DOI] [PubMed] [Google Scholar]
- 24.Barrile R, van der Meer AD, Park H, Fraser JP, Simic D, Teng F, et al. Organ-on-chip recapitulates thrombosis induced by an anti-CD154 monoclonal antibody: translational potential of advanced microengineered systems. Clin Pharmacol Ther. Epub ahead of print 27 February 2018. DOI: 10.1002/cpt.1054. [DOI] [PubMed] [Google Scholar]
- 25.Passini E, Britton OJ, Lu HR, Rohrbacher J, Hermans AN, Gallacher DJ, et al. Human in silico drug trials demonstrate higher accuracy than animal models in predicting clinical pro-arrhythmic cardiotoxicity. Front Physiol 2017; 8: 668–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nguyen N, Nguyen W, Nguyenton B, Ratchada P, Page G, Miller PE, et al. Adult human primary cardiomyocyte-based model for the simultaneous prediction of drug-induced inotropic and pro-arrhythmia risk. Front Physiol 2017; 8: 1073–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Thompson RA, Isin EM, Li Y, Weidolf L, Page K, Wilson I, et al. In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs. Chem Res Toxicol 2012; 25: 1616–1632. [DOI] [PubMed] [Google Scholar]
- 28.Kenna JG, Stahl SH, Eakins JA, Foster AJ, Andersson LC, Bergare J, et al. Multiple compound-related adverse properties contribute to liver injury caused by endothelin receptor antagonists. J Pharmacol Exp Ther 2015; 352: 281–290. [DOI] [PubMed] [Google Scholar]
- 29.Ware BR, Berger DR, Khetani SR. Prediction of drug-induced liver injury in micropatterned co-cultures containing iPSC-derived human hepatocytes. Toxicol Sci 2015; 145: 252–262. [DOI] [PubMed] [Google Scholar]
- 30.Proctor WR, Foster AJ, Vogt J, Summers C, Middleton B, Pilling MA, et al. Utility of spherical human liver microtissues for prediction of clinical drug-induced liver injury. Arch Toxicol 2017; 91: 2849–2863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Garside H, Marcoe KF, Chesnut-Speelman J, Foster AJ, Muthas D, Kenna JG, et al. Evaluation of the use of imaging parameters for the detection of compound-induced hepatotoxicity in 384-well cultures of HepG2 cells and cryopreserved primary human hepatocytes. Toxicol In Vitro 2014; 28: 171–181. [DOI] [PubMed] [Google Scholar]
- 32.Woodhead JL, Watkins PB, Howell BA, Siler SQ, Shoda LK. The role of quantitative systems pharmacology modeling in the prediction and explanation of idiosyncratic drug-induced liver injury. Drug Metab Pharmacokinet 2017; 32: 40–45. [DOI] [PubMed] [Google Scholar]
- 33.Wallis R, Benson C, Darpo B, Gintant G, Kanda Y, Prasad K, et al. CiPA challenges and opportunities from a non-clinical, clinical and regulatory perspectives. An overview of the safety pharmacology scientific discussion. J Pharmacol Toxicol Methods 2018; 93: 15–25. [DOI] [PubMed] [Google Scholar]
- 34.Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Human iPSC-derived neuronal models for in vitro neurotoxicity assessment. Neurotoxicology 2018; 67: 215–225. [DOI] [PubMed] [Google Scholar]
- 35.Bavli D, Prill S, Ezra E, Levy G, Cohen M, Vinken M, et al. Real-time monitoring of metabolic function in liver-on-chip microdevices tracks the dynamics of mitochondrial dysfunction. Proc Natl Acad Sci U S A 2016; 113: E2231–E2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Prill S, Bavli D, Levy G, Ezra E, Schmälzlin E, Jaeger MS, et al. Real-time monitoring of oxygen uptake in hepatic bioreactor shows CYP450-independent mitochondrial toxicity of acetaminophen and amiodarone. Arch Toxicol 2016; 90: 1181–1191. [DOI] [PubMed] [Google Scholar]
- 37.van Esbroeck AC, Janssen AP, Cognetta AB, Ogasawara D, Shpak G, van der Kroeg M, et al. Activity-based protein profiling reveals off-target proteins of the FAAH inhibitor BIA 10-2474. Science 2017; 356: 1084–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dirven H, Vist G, Sayed A, Bandhakavi S, Branik M, Peace N, et al. Troglitazone and Rosiglitazone profiles in Tox21/Toxcast: verifying results with toxicology data from published in vivo data and human pharmacovigilance data using an evidence-based approach. Toxicol Lett 2018; 295S: S56–S68. [Google Scholar]
- 39.Hartung T. Fundamentals of an evidence-based toxicology. Hum Exp Toxicol 2009; 28: 93–94. [DOI] [PubMed] [Google Scholar]
- 40.Birnbaum LS, Thayer KA, Bucher JR, Wolfe MS. Implementing systematic review at the National Toxicology Program: status and next steps. Environ Health Perspect 2013; 121: a108–a108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.OECD (Organisation for Economic Co-operation and Development). Integrated approaches to testing and assessment (IATA), www.oecd.org/chemicalsafety/risk-assessment/iata-integrated-approaches-to-testing-and-assessment.htm (2018, accessed 27 April 2018).
- 42.Casati S. Integrated approaches to testing and assessment. Basic Clin Pharmacol Toxicol 2018; 123: 51–55. [DOI] [PubMed] [Google Scholar]
- 43.Luechtefeld T, Marsh D, Rowlands C, Hartung T. Machine learning of toxicological big data enables read-across structure activity relationships (RASAR) outperforming animal test reproducibility. Toxicol Sci 2018; 165: 198–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Burt T, Yoshida K, Lappin G, Vuong L, John C, de Wildt SN, et al. Microdosing and other phase 0 clinical trials: facilitating translation in drug development. Clin Transl Sci 2016; 9: 74–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Arnaud C. In the search for new drugs, diverging roads for microdosing. Chem Eng News 2013; 91: 9–13. [Google Scholar]
- 46.Kenna JG. Human biology-based drug safety evaluation: scientific rationale, current status and future challenges. Expert Opin Drug Metab Toxicol 2017; 13: 567–574. [DOI] [PubMed] [Google Scholar]
- 47.Heneghan C, Goldacre B, Mahtani KR. Why clinical trial outcomes fail to translate into benefits for patients. Trials 2017; 18: 122–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Earley A, Lau J, Uhlig K. Haphazard reporting of deaths in clinical trials: a review of cases of ClinicalTrials.gov records and matched publications – a cross-sectional study. BMJ Open 2013; 3: e001963–e001963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Tan DS, Thomas GV, Garrett MD, Banerji U, de Bono JS, Kaye SB, et al. Biomarker-driven early clinical trials in oncology: a paradigm shift in drug development. Cancer J 2009; 15: 406–420. [DOI] [PubMed] [Google Scholar]
- 50.Dienstmann R, Rodon J, Tabernero J. Biomarker-driven patient selection for early clinical trials. Curr Opin Oncol 2013; 25: 305–312. [DOI] [PubMed] [Google Scholar]
- 51.Hartung T. On validation. ALTEX 2007; 24: 67–80. [DOI] [PubMed] [Google Scholar]
- 52.Leist M, Hasiwa N, Daneshian M, Hartung T. Validation and quality control of replacement alternatives–current status and future challenges. Toxicol Res 2012; 1: 8–22. [Google Scholar]
- 53.Sewell F, Doe J, Gellatly N, Ragan I, Burden N. Steps towards the international regulatory acceptance of non-animal methodology in safety assessment. Regul Toxicol Pharmacol 2017; 89: 50–56. [DOI] [PubMed] [Google Scholar]
- 54.Food and Drug Administration. FDA’s predictive toxicology roadmap, www.fda.gov/downloads/ScienceResearch/SpecialTopics/…/UCM587831.pdf (2017, accessed 26 April 2018).
- 55.Interagency Coordinating Committee on the Validation of Alternative Methods. A strategic roadmap for establishing new approaches to evaluate the safety of chemicals and medical products in the United States, https://ntp.niehs.nih.gov/pubhealth/evalatm/natl-strategy/index.html (2018, accessed 26 April 2018).
- 56.Casey W. A strategic roadmap for establishing new approaches to evaluate the safety of chemicals and medical products in the United States. Conference paper, John Hopkins, Bloomberg School of Public Health, www.dropbox.com/s/3nrphg4aqdehf44/2018-05-30_EHE_Becton.mp4?dl=0 (2018, accessed 18 July 18).
- 57.Frank J. Technological lock-in, positive institutional feedback, and research on laboratory animals. Struct Change Econ Dynam 2005; 16: 557–575. [Google Scholar]
- 58.Geels FW. The multi-level perspective on sustainability transitions: responses to seven criticisms. Environ Innov Soc Transit 2011; 1: 24–40. [Google Scholar]
- 59.Seok J, Warren HS, Cuenca AG, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci 2013; 110: 3507–3512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Konomi K, Tobita M, Kimura K, Sato D. New Japanese initiatives on stem cell therapies. Cell Stem Cell 2015; 16: 350–352. [DOI] [PubMed] [Google Scholar]