Guest Column | January 8, 2024

Animal Testing Isn't Perfect. Is There An Alternative?

By Anke Rosch, Boehringer Ingelheim

Rat-GettyImages-680037876

When I started my career in drug safety, friends warned me that toxicology is as old as humanity. This would also apply to its methods: many animal experiments, regulations, and a traditional mindset, making the integration of innovative (animal-free) methods impossible. I would like to clear up these outdated prejudices.

Animal Statistics

In the European Union, 10.4 million animals were used for the first time in research and testing in 2019 (down 1.5% vs. 2018), including approximately 6.4 million rodent and 700,000 non-rodent mammals.1 For the United States, no easily accessible data on animal use is available. The United States Department of Agriculture (USDA) reported around 800,000 (up 2.2% vs. 2018)2 laboratory animals used for research under the Animal Welfare Act (AWA)3 in 2019, excluding purpose-bred mice and rats. Carbone4 presented an annual estimate of 111.5 million rodents about the same time, based on data from 16 large American institutions. However, in Europe, only 7% of the total number of animals participated in toxicity and safety testing (down 2% vs. 2018)1 compared to 72% used for research.

Regulatory Considerations

In 1937, mass poisoning in the United States because of the antibiotic Elixir Sulfanilamid (in diethylene glycol) led to the passage of the Food, Drug, and Cosmetic Act in 1938.5 The sedative Contergan (thalidomide),6 dosed to pregnant women, caused severe birth defects and deaths in thousands of children in the 1950s. As a result, the Kefauver-Harris amendments were enacted in 1962, requiring animal testing for drug safety and efficacy. Today, several guidelines thoroughly regulate preclinical drug development, such as ICH M3(R2)7 for human pharmaceuticals, ICH S6R1 for biotechnology-derived products,8 ICH S9 for anticancer pharmaceuticals,9 and ICH S5(R3) for reproductive and developmental toxicity.10

The high attrition rate of new drugs (90%)11 after entering clinical studies despite animal testing led to the release of the FDA Modernization Act 2.0 in 2022.12 This law permits the use of suitable animal-free new approach methodologies (NAMs) instead of mandatory animal studies. Simultaneously, the European Parliament launched a program to eliminate animal studies "as soon as scientifically possible," inspired by a European citizen initiative13 in 2021.14 The reformation of the European Directive 2010/63/EU15,16 on the Protection of Animals for Scientific Purpose is supported by the European Medicines Agency (EMA) and the European Partnership for Alternative Approaches to Animal Testing (EPAA).

Reasons For A Change

The heart of the debate about animal sacrifice for drug development lies in the question of whether animal lives are less valuable than those of humans.

There are many additional reasons to dismiss animal studies. Rodent and non-rodent experiments predicted human toxicity in only 48%17 to 71%18 of drugs. In about 20% of drugs 19 like antibodies and fusion and recombinant proteins, the human-specific mode did not allow extrapolation from animal to potential human immunogenicity. In 2006, six human volunteers had to experience that firsthand by participating in a Phase 1 clinical trial with the CD28 superagonist antibody TGN1412.20 It caused systemic "cytokine storm" and life‑threatening multiorgan failure, even though it had been tested previously in two animal species.21 Forty-five percent11 of novel drug failures in clinical studies are attributed to unanticipated toxicity.22 Only 19% of human serious adverse events post marketing were detected in animals beforehand.22

On the contrary, toxicity in animals is occasionally not predictive for humans and might have prevented approval of promising drugs. Liver and kidney toxicity in cats and dogs or embryo toxicity in rats and monkeys, for instance, would have probably discontinued the pharmaceutical development of paracetamol23 and aspirin,24 respectively.

Paradigm Shift In Preclinical Safety

Carcinogenicity may serve as an example of systemic toxicology testing with high complexity. Therefore, the use of alternative methods is still limited. For genotoxic carcinogens, the guidelines include a standard battery of tests, i.e., the Ames test, the micronucleus test, and the chromosomal aberration assay performed in-vitro or using material from repeated dose animal studies since 1995.25–28

For in-vivo carcinogenicity testing with up to 600 to 800 animals per study,29 the ICH S1B addendum30 released in 2022 describes a novel tiered testing approach. Previous versions requested a two-year (lifetime) carcinogenicity study in rats in association with a two-year or medium-term (six months) study in a second species (mouse or transgenic rasH2 mice, respectively). The addendum now allows omitting the two-year rat carcinogenicity study based on available pharmacological and toxicological data in a weight-of-evidence (WoE) approach. Additionally, regulators enable the implementation of mechanistic in-vitro data. 

NAMs for non-genotoxic carcinogen testing such as the cell transformation assay31 or a genomic biomarker panel32 to predict rodent carcinogenicity are validated by an OECD established expert group33–35 for regulatory integration.36

From 2015 to 2019, FDA approved new drug applications based on 109 carcinogenicity studies, including at least 65,341 rats and mice. The novel approach promises a 46% reduction in this animal number.37

In the 1980s, several FDA approved drugs (Seldane/terfenadine and Hismanal/astemizole)38 made it into the headlines when a potentially fatal polymorphic ventricular arrhythmia (Torsade de Pointes) occurred in humans. The delay in cardiac repolarization by inhibition of the rapidly activating delayed rectifier potassium current Ikr or hERG (human-ether-a-go-go) channel associated with prolongation of the QT interval was held responsible. As a result, the Committee for Proprietary Medicinal Products (CPMP) published an early guideline on the evaluation of ventricular repolarization39,40 in 1997. The more detailed ICH S7B guidance41 released in 2005 declared a preclinical in-vitro hERG and an in-vivo QT assay, such as in telemetrized animals, as mandatory.

One hundred percent of the responders from surveyed pharmaceutical companies performed in-vivo telemetry studies42 in the following years. Twenty-six percent of the responders even implemented proarrhythmia models such as the methoxamine-treated rabbit and the chronically atrioventricular blocked dog42 that were, however, totally rejected by 62% of scientists. In 2013, a "think tank"43 sponsored by the Cardiac Safety Research Consortium (CSRC), Health and Environmental Sciences Institute (HESI), and FDA confirmed that Ikr inhibition is not the only reason for Torsade de Pointes. A 3R-compliant new testing strategy development was started by the comprehensive in-vitro proarrhythmia assay (CiPa) initiative for updating the S7B and E14 guidelines. The ICH E14/S7B Q&As44 were released in 2022 with the primary aim of waiving thorough QT studies by providing additional preclinical information without increasing the use of animals for drug development. Two years before release, the number of stand-alone telemetry studies had decreased to 77% and 70% using dogs and minipigs or monkeys ("frequently and always"),45 respectively, presumably for the benefit of jacket telemetry measurements in toxicity studies.

Nowadays, preclinical data supplementing the hERG and in vivo assay will consist of animal‑free methods such as the assessment of drug effects on multiple ion channels46 with subsequent in-silico modeling47,48 by the updated O'Hara–Rudy cell model and on human iPSC-derived cardiomyocytes.49 Furthermore, it is recommended to perform a more detailed investigation of drug effects already in Phase 1 studies associated with plasma concentration-QTc modeling.50

Conclusion

I agree that animal experiments cannot entirely predict human outcomes in clinical studies. Differences in metabolic pathways, toxicokinetic profile, drug metabolites, receptor distribution, and sensitivity to adverse effects are responsible. However, often the focus is rather on the failures of animal studies and not on the potential harm that humans could have faced if one hadn't conducted them. For drug safety, guidelines restrict flexibility and extend timelines for drug development but ensure well-powered studies with controlled animal numbers. Reasons for increased animal usage in research may lie in a lack of harmonization. Multiple scientific groups all over the world work competitively on the same promising topic with divergent and often underpowered studies. A systematic meta-analysis of results could reduce the need for animal testing.

Despite the existence of guidelines for safety testing, scientists and regulators have not hesitated to initiate a paradigm shift to bring toxicology into the 21st century. NAMs are stepwise taken up into regulatory testing strategies following validation against animal studies. The mindset of toxicologists got an upgrade with technologies such as adverse outcome pathways (AoPs),51 read-across,52 microphysiological systems (organ-on-a-chip and 3D organ constructs),53 and artificial intelligence.54 However, a biological system is not only the sum of its functions but also of synergies among them.

Replacing animals entirely in toxicology studies by in-vitro methods still seems too risky because it oversimplifies their complexity. I strongly believe that we can achieve further reduction of animal numbers in research and development. I regret having to say that we probably must perform a certain defined number of animal studies for the investigations of adverse effects resulting from interaction between different organs and physiological systems. Integration of further endpoints will significantly refine these studies and make other studies obsolete. Supplementary NAMs might reduce their number as well. The challenge is bridging the gap between traditional and modern toxicologists to benefit from each other's expertise for the animals' sake.

References

  1. European Commission Summary report on the statistics on the use of animals for scientific purposes in the Member States of the European Union and Norway in 2019. SWD(2022) 199. 2022. Part 1/2 (EU statistics).
  2. United States Department of Agriculture, Animal and Plant Health Inspection Service. Annual Report Animal Usage by Fiscal Year. Total number of animal research facilities in regulated activities (Column B). 2019. united-states-2019.pdf (wordpress.com) Accessed on 12 Dec 2023.
  3. The Animal Welfare Act: Background and Selected Issues. 2023. R47179 (congress.gov) R47179 (congress.gov) Accessed on 12 Dec 2023.
  4. Carbone, L. Estimating mouse and rat use in American laboratories by extrapolation from Animal Welfare Act-regulated species. Sci Rep 11, 493 (2021). https://doi.org/10.1038/s41598-020-79961-0.
  5. Wax PM. Elixirs, diluents, and the passage of the 1938 Federal Food, Drug and Cosmetic act. Annals of internal medicine. 1995 Mar 15;122(6):456-461. doi: 10.7326/0003-4819-122-6-199503150-00009.
  6. Kim JH, Scialli AR. Thalidomide: the tragedy of birth defects and the effective treatment of disease. Toxicol Sci. 2011 Jul;122(1):1-6. doi: 10.1093/toxsci/kfr088. Epub 2011 Apr 19. Erratum in: Toxicol Sci. 2012 Feb;125(2):613. PMID: 21507989.
  7. EMA. ICH M3(R2). ICH Guideline M3(R2) on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals. Step 5. EMA/CPMP/ICH/286/1995. 2009. M3(R2) Step 5 Non-clinical safety studies for conduct of human clinical trials for pharmaceuticals (europa.eu). Accessed on 12 Dec 2023.
  8. EMA. ICH S6(R1). Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals. Step 5. EMA/CHMP/ICH/731268/1998. 2011 ICH S6 (R1) Preclinical safety evaluation of biotechnology-derived pharmaceuticals - Scientific guideline | European Medicines Agency (europa.eu). Accessed on 12 Dec 2023.
  9. EMA. ICH S9. Nonclinical Evaluation for Anticancer Pharmaceuticals. Step 5. EMA/CHMP/ICH/646107/2008. 2009. ICH S9 Non-clinical evaluation for anticancer pharmaceuticals - Scientific guideline | European Medicines Agency (europa.eu) Accessed on 12 Dec 2023.
  10. EMA. ICH S5(R3). International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). Detection of Reproductive and Developmental Toxicity for Human Pharmaceuticals. 2020. S5-R3_Step4_Guideline_2020_0218_1.pdf (ich.org). Accessed on 12 Dec 2023.
  11. Sun D, Gao W, Hu H, Zhou S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm Sin B. 2022 Jul;12(7):3049-3062. doi: 10.1016/j.apsb.2022.02.002. Epub 2022 Feb 11. PMID: 35865092; PMCID: PMC9293739.
  12. CONGRESS.GOV. H.R.2617 - Consolidated Appropriations Act, 2023. Public Law No: 117-328. December 29, 2022. S.5002 - 117th Congress (2021-2022): FDA Modernization Act 2.0 | Congress.gov | Library of Congress. Accessed 12 Dec 2023.
  13. Save Cruelty Free Cosmetics-Commit to a Europe without animal testing.: Brussels, Belgium, 2021. Save Cruelty Free Cosmetics - Commit to a Europe Without Animal Testing (europa.eu) Accessed on 12 Dec 2023.
  14. European Parliament. Plans and Actions to Accelerate a Transition to Innovation without the Use of Animals in Research, Regulatory Testing and Education; European Parliament: Strasbourg, France, 2021. MEPs demand EU action plan to end the use of animals in research and testing | News | European Parliament (europa.eu). Accessed on 12 Dec 2023.
  15. European Commission. Directive 2010/63/Eu of the European Parliament and of the Council of 22 September 2010 on the Protection of Animals Used for Scientific Purposes. In Directive 63/2010/EU; European Commission: Brussels, Belgium, 2010. EUR-Lex - 32010L0063 - EN - EUR-Lex (europa.eu). Accessed on 12 Dec 2023.
  16. Marshall LJ, Constantino H, Seidle T. Phase-In to Phase-Out-Targeted, Inclusive Strategies Are Needed to Enable Full Replacement of Animal Use in the European Union. Animals (Basel). 2022 Mar 29;12(7):863. doi: 10.3390/ani12070863. PMID: 35405853; PMCID: PMC8997151.
  17. Tamaki C, Nagayama T, Hashiba M, Fujiyoshi M, Hizue M, Kodaira H, Nishida M, Suzuki K, Takashima Y, Ogino Y, Yasugi D, Yoneta Y, Hisada S, Ohkura T, Nakamura K. Potentials and limitations of nonclinical safety assessment for predicting clinical adverse drug reactions: correlation analysis of 142 approved drugs in Japan. J Toxicol Sci. 2013;38(4):581-98. doi: 10.2131/jts.38.581. PMID: 23824014.
  18. 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. Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol. 2000 Aug;32(1):56-67. doi: 10.1006/rtph.2000.1399. PMID: 11029269.
  19. American Pharmaceutical Review. Preclinical Development: The Safety Hurdle Prior to Human Trials. American Pharmaceutical Review. April 30, 2016. Available at: Preclinical Development: The Safety Hurdle Prior to Human Trials | American Pharmaceutical Review - The Review of American Pharmaceutical Business & Technology. Accessed 12 Dec 2023.
  20. Attarwala H. TGN1412: From Discovery to Disaster. J Young Pharm. 2010 Jul;2(3):332-6. doi: 10.4103/0975-1483.66810. PMID: 21042496; PMCID: PMC2964774.
  21. Horvath C, Andrews L, Baumann A, Black L, Blanset D, Cavagnaro J, Hastings KL, Hutto DL, MacLachlan TK, Milton M, Reynolds T, Roberts S, Rogge M, Sims J, Treacy G, Warner G, Green JD. Storm forecasting: additional lessons from the CD28 superagonist TGN1412 trial. Nat Rev Immunol. 2012 Oct;12(10):740; author reply 740. doi: 10.1038/nri3192-c1. Epub 2012 Aug 31. Erratum in: Nat Rev Immunol. 2012 Oct;12(10):740. PMID: 22941443.
  22. van Meer PJ, Kooijman M, Gispen-de Wied CC, Moors EH, Schellekens H. The ability of animal studies to detect serious post marketing adverse events is limited. Regul Toxicol Pharmacol. 2012 Dec;64(3):345-9. doi: 10.1016/j.yrtph.2012.09.002. Epub 2012 Sep 12. PMID: 22982732.
  23. Villar D, Buck WB, Gonzalez JM. Ibuprofen, aspirin and acetaminophen toxicosis and treatment in dogs and cats. Vet Hum Toxicol. 1998 Jun;40(3):156-62. PMID: 9610496.
  24. Wilson JG, Ritter EJ, Scott WJ, Fradkin R. Comparative distribution and embryotoxicity of acetylsalicylic acid in pregnant rats and rhesus monkeys. Toxicol Appl Pharmacol. 1977 Jul;41(1):67-78. doi: 10.1016/0041-008x(77)90054-0. PMID: 408942.
  25. Müller L, Kikuchi Y, Probst G, Schechtman L, Shimada H, Sofuni T, Tweats D. ICH-harmonised guidances on genotoxicity testing of pharmaceuticals: evolution, reasoning and impact. Mutat Res. 1999 May;436(3):195-225. doi: 10.1016/s1383-5742(99)00004-6. PMID: 10354523.
  26. EMA. ICH S2A: Note for Guidance on Genotoxicity: Specific Aspects of Regulatory Genotoxicity Tests for Pharmaceuticals. CPMP/ICH/141/95. 1995. S 2 A Genotoxicity: Specific Aspects of Regulatory Genotoxicity Tests for Pharmaceuticals (europa.eu). Accessed on 12 Dec 2023.
  27. EMA. ICH S2B: Note for Guidance on genotoxicity: A Standard Battery for Genotoxicity Testing of Pharmaceuticals. Step 5, CPMP/ICH/174/95. 1997. S 2 B Genotoxicity: A Standard Battery for Genotoxicity Testing of Pharmaceuticals (europa.eu). Accessed on 12 Dec 2023.
  28. EMA. ICH S2(R1) ICH Guideline S2 (R1) on genotoxicity testing and data interpretation for pharmaceuticals intended for human use. ICH S2 (R1) Genotoxicity testing and data interpretation for pharmaceuticals intended for human use - Scientific guideline | European Medicines Agency (europa.eu) Step 5, EMA/CHMP/ICH/126642. 2011. Accessed on 12 Dec 2023.
  29. Basketter DA, Clewell H, Kimber I, Rossi A, Blaauboer B, Burrier R, Daneshian M, Eskes C, Goldberg A, Hasiwa N, Hoffmann S, Jaworska J, Knudsen TB, Landsiedel R, Leist M, Locke P, Maxwell G, McKim J, McVey EA, Ouédraogo G, Patlewicz G, Pelkonen O, Roggen E, Rovida C, Ruhdel I, Schwarz M, Schepky A, Schoeters G, Skinner N, Trentz K, Turner M, Vanparys P, Yager J, Zurlo J, Hartung T. A roadmap for the development of alternative (non-animal) methods for systemic toxicity testing. ALTEX. 2012;29(1):3-91. doi: 10.14573/altex.2012.1.003. PMID: 22307314.
  30. EMA. ICH S1B(R1). ICH Guideline S1B(R1) on testing for carcinogenicity of pharmaceuticals, Step 5. EMA/774371/2022. ICH guideline S1B(R1) on testing for carcinogenicity of pharmaceuticals Step 5 (europa.eu). Accessed on 12 Dec 2023.
  31. Stuart Creton, Marilyn J. Aardema, Paul L. Carmichael, James S. Harvey, Francis L. Martin, Robert F. Newbold, Michael R. O’Donovan, Kamala Pant, Albrecht Poth, Ayako Sakai, Kiyoshi Sasaki, Andrew D. Scott, Leonard M. Schechtman, Rhine R. Shen, Noriho Tanaka, Hemad Yasaei, Cell transformation assays for prediction of carcinogenic potential: state of the science and future research needs, Mutagenesis, Volume 27, Issue 1, January 2012, Pages 93–101, https://doi.org/10.1093/mutage/ger053. Accessed on 12 Dec 2023.
  32. Corton JC, Mitchell CA, Auerbach S, Bushel P, Ellinger-Ziegelbauer H, Escobar PA, Froetschl R, Harrill AH, Johnson K, Klaunig JE, Pandiri AR, Podtelezhnikov AA, Rager JE, Tanis KQ, van der Laan JW, Vespa A, Yauk CL, Pettit SD, Sistare FD. A Collaborative Initiative to Establish Genomic Biomarkers for Assessing Tumorigenic Potential to Reduce Reliance on Conventional Rodent Carcinogenicity Studies. Toxicol Sci. 2022 Jun 28;188(1):4-16. doi: 10.1093/toxsci/kfac041. PMID: 35404422; PMCID: PMC9238304.
  33. Jacobs MN, Colacci A, Corvi R, Vaccari M, Aguila MC, Corvaro M, Delrue N, Desaulniers D, Ertych N, Jacobs A, Luijten M, Madia F, Nishikawa A, Ogawa K, Ohmori K, Paparella M, Sharma AK, Vasseur P. Chemical carcinogen safety testing: OECD expert group international consensus on the development of an integrated approach for the testing and assessment of chemical non-genotoxic carcinogens. Arch Toxicol. 2020 Aug;94(8):2899-2923. doi: 10.1007/s00204-020-02784-5. Epub 2020 Jun 27. PMID: 32594184; PMCID: PMC7395040.
  34. OECD, 2017. Guidance Document on the In Vitro Bhas 42 Cell Transformation Assay (BHAS 42 CTA). OECD Environment, Health and Safety Publications Series on Testing and Assessment No. 231. ENV/JM/MONO(2016)1. Bhas 42 CTA GD after 3rd comments-F_CLEAN.pdf (oecd.org). Accessed 12 Dec 2023.
  35. OECD, 2015. Guidance Document on the In Vitro Syrian Hamster Embryo (SHE) Cell Transformation Assay. OECD Environment, Health and Safety Publications Series on Testing and Assessment No. 214. ENV/JM/MONO(2015)18 Guidance-Document-on-the-in-vitro-Syrian-Hamster-Embryo-Cell-Transformation-Assay.pdf (oecd.org). Accessed 12 Dec 2023.
  36. van der Laan JW, Andersson M, Beken S, Bonelli M, Brendler-Schwaab S, Kane R, Pasanen M, Ponzano S, Paur J, Siezen C, Soleng A, Whomsley R. EMA commentary on the ICH guideline for testing for carcinogenicity of pharmaceuticals. Br J Clin Pharmacol. 2023 Aug;89(8):2341-2348. doi: 10.1111/bcp.15790. Epub 2023 Jun 2. PMID: 37265032.
  37. Manuppello J, Slankster-Schmierer E, Baker E, Sullivan K. Animal use and opportunities for reduction in carcinogenicity studies supporting approved new drug applications in the U.S., 2015-2019. Regul Toxicol Pharmacol. 2023 Jan;137:105289. doi: 10.1016/j.yrtph.2022.105289. Epub 2022 Nov 12. PMID: 36379352.
  38. Li M, Ramos LG. Drug-Induced QT Prolongation And Torsades de Pointes. P T. 2017 Jul;42(7):473-477. PMID: 28674475; PMCID: PMC5481298.
  39. Crumb W, Cavero I. QT interval prolongation by non-cardiovascular drugs: issues and solutions for novel drug development. Pharm Sci Technol Today. 1999 Jul;2(7):270-280. doi: 10.1016/s1461-5347(99)00172-8. PMID: 10407390.
  40. Points to consider CPMP QT prolongation Committee for Proprietary Medicinal Products (1997) CPMP/986/96. annual-report-european-agency-evaluation-medicinal-products-1997_en.pdf (europa.eu) Accessed on 12 Dec 2023.
  41. EMA. ICH S7B. The Nonclincial Evaluation of the potential for delayed ventricular repolarization (QT Interval Prolongation) by Human Pharmaceuticals. Step 5. CPMP/ICH/423/02. 2005. S 7 B The nonclinical Evaluation of the Potential for delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceuticals (europa.eu) accessed on 12 Dec 2023.
  42. Lindgren S, Bass AS, Briscoe R, Bruse K, Friedrichs GS, Kallman MJ, Markgraf C, Patmore L, Pugsley MK. Benchmarking safety pharmacology regulatory packages and best practice. J Pharmacol Toxicol Methods. 2008 Sep-Oct;58(2):99-109. doi: 10.1016/j.vascn.2008.07.001. Epub 2008 Jul 19. PMID: 18692579.
  43. Sager PT, Gintant G, Turner JR, Pettit S, Stockbridge N. Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. Am Heart J. 2014 Mar;167(3):292-300. doi: 10.1016/j.ahj.2013.11.004. Epub 2013 Dec 2. PMID: 24576511.
  44. EMA. ICH Guideline E14/S7B: Clinical and Nonclincial Evaluation of QT/QTc Interval Prolongation and Proarrhythmic Potential-questions and answers. Step 5. EMA/CHMP/ICH/415588/2020. 2022. ICH guideline E14_S7B on clinical and nonclinical evaluation of QT_QTc interval prolongation and proarrhythmic potential- questions & answers Step 5 (europa.eu). Accessed on 12 Dec 2023.
  45. Authier S, Abernathy MM, Correll K, Chui RW, Dalton J, Foley CM, Friedrichs GS, Koerner JE, Kallman MJ, Pannirselvam M, Redfern WS, Urmaliya V, Valentin JP, Wisialowski T, Zabka TS, Pugsley MK. An Industry Survey With Focus on Cardiovascular Safety Pharmacology Study Design and Data Interpretation. Int J Toxicol. 2020 Jul/Aug;39(4):274-293. doi: 10.1177/1091581820921338. Epub 2020 May 14. PMID: 32406289.
  46. Li Z, Dutta S, Sheng J, Tran PN, Wu W, Chang K, Mdluli T, Strauss DG, Colatsky T. Improving the In Silico Assessment of Proarrhythmia Risk by Combining hERG (Human Ether-à-go-go-Related Gene) Channel-Drug Binding Kinetics and Multichannel Pharmacology. Circ Arrhythm Electrophysiol. 2017 Feb;10(2):e004628. doi: 10.1161/CIRCEP.116.004628. Erratum in: Circ Arrhythm Electrophysiol. 2017 Mar;10 (3): PMID: 28202629.
  47. O'Hara T, Virág L, Varró A, Rudy Y. Simulation of the undiseased human cardiac ventricular action potential: model formulation and experimental validation. PLoS Comput. Biol. 2011 May;7(5):e1002061. doi: 10.1371/journal.pcbi.1002061. Epub 2011 May 26. PMID: 21637795; PMCID: PMC3102752.
  48. Dutta S, Chang KC, Beattie KA, Sheng J, Tran PN, Wu WW, Wu M, Strauss DG, Colatsky T, Li Z. Optimization of an In silico Cardiac Cell Model for Proarrhythmia Risk Assessment. Front Physiol. 2017 Aug 23;8:616. doi: 10.3389/fphys.2017.00616. Erratum in: Front Physiol. 2017 Dec 06;8:1025. PMID: 28878692; PMCID: PMC5572155.
  49. Blinova K, Dang Q, Millard D, Smith G, Pierson J, Guo L, Brock M, Lu HR, Kraushaar U, Zeng H, Shi H, Zhang X, Sawada K, Osada T, Kanda Y, Sekino Y, Pang L, Feaster TK, Kettenhofen R, Stockbridge N, Strauss DG, Gintant G. International Multisite Study of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Drug Proarrhythmic Potential Assessment. Cell Rep. 2018 Sep 25;24(13):3582-3592. doi: 10.1016/j.celrep.2018.08.079. PMID: 30257217; PMCID: PMC6226030.
  50. Darpo B, Ferber G. The New S7B/E14 Question and Answer Draft Guidance for Industry: Contents and Commentary. J Clin Pharmacol. 2021 Oct;61(10):1261-1273. doi: 10.1002/jcph.1880. Epub 2021 Jun 8. PMID: 33896027; PMCID: PMC9290990.
  51. Chauhan V, Hamada N, Wilkins R, Garnier-Laplace J, Laurier D, Beaton D, Tollefsen KE. A high-level overview of the OECD AOP Development Programme. Int J Radiat Biol. 2022;98(12):1704-1713. doi: 10.1080/09553002.2022.2110311. Epub 2022 Aug 24. PMID: 35938955.
  52. Schultz TW, Amcoff P, Berggren E, Gautier F, Klaric M, Knight DJ, Mahony C, Schwarz M, White A, Cronin MT. A strategy for structuring and reporting a read-across prediction of toxicity. Regul Toxicol Pharmacol. 2015 Aug;72(3):586-601. doi: 10.1016/j.yrtph.2015.05.016. Epub 2015 May 21. PMID: 26003513.
  53. Rothbauer M, Bachmann BEM, Eilenberger C, Kratz SRA, Spitz S, Höll G, Ertl P. A Decade of Organs-on-a-Chip Emulating Human Physiology at the Microscale: A Critical Status Report on Progress in Toxicology and Pharmacology. Micromachines (Basel). 2021 Apr 21;12(5):470. doi: 10.3390/mi12050470. PMID: 33919242; PMCID: PMC8143089.
  54. Lin Z, Chou WC. Machine Learning and Artificial Intelligence in Toxicological Sciences. Toxicol Sci. 2022 Aug 25;189(1):7-19. doi: 10.1093/toxsci/kfac075. PMID: 35861448; PMCID: PMC9609874.

Anke Rosch is a board-certified pharmacologist and toxicologist working at Boehringer Ingelheim Pharma GmbH & Co. KG. A doctor of veterinary medicine, she has more than 20 years of experience in the pharmaceutical industry and has special expertise in safety pharmacology. Anke can be reached at ankerosch.PharmacolTox@t-online.de.