Abstract
Drug-induced kidney injury (DIKI) is a frequently reported adverse event, associated with acute kidney injury, chronic kidney disease, and end-stage renal failure. Prospective cohort studies on acute injuries suggest a frequency of around 14%–26% in adult populations and a significant concern in pediatrics with a frequency of 16% being attributed to a drug. In drug discovery and development, renal injury accounts for 8 and 9% of preclinical and clinical failures, respectively, impacting multiple therapeutic areas. Currently, the standard biomarkers for identifying DIKI are serum creatinine and blood urea nitrogen. However, both markers lack the sensitivity and specificity to detect nephrotoxicity prior to a significant loss of renal function. Consequently, there is a pressing need for the development of alternative methods to reliably predict drug-induced kidney injury (DIKI) in early drug discovery. In this article, we discuss various aspects of DIKI and how it is assessed in preclinical models and in the clinical setting, including the challenges posed by translating animal data to humans. We then examine the urinary biomarkers accepted by both the US Food and Drug Administration (FDA) and the European Medicines Agency for monitoring DIKI in preclinical studies and on a case-by-case basis in clinical trials. We also review new approach methodologies (NAMs) and how they may assist in developing novel biomarkers for DIKI that can be used earlier in drug discovery and development.
Keywords: renal injury, drug discovery, nephrotoxicity, DIKI, kidney injury
Introduction
Kidney disease is the 10th leading cause of death in the United States (U.S.), contributing to 54,358 deaths in 2021 alone, with nephritis, nephrotic syndrome, and nephrosis accounting for this mortality.1 While nephrotoxicity comprises a wide spectrum of diseases, here we define it as the rapid deterioration of kidney function or kidney injury due to the damaging and toxic effects of drugs, chemicals, and toxins.2 Around 20% of nephrotoxicity cases are attributed to drugs.2 Drug-induced kidney injury (DIKI) can lead to the development of acute kidney injury (AKI), chronic kidney disease (CKD), or end-stage renal disease, causing over 1.5 million adverse events annually and affecting approximately 26% of the U.S. population.3,4 Prospective cohort studies on acute injury suggest a frequency of around 14%–26% in adult populations and a significant concern in pediatrics, where 16% of acute injury cases are attributed to drug-induced causes (see Awdishu and Mehta 20175 for a detailed review of incidence).
In addition to the impact on patients and the healthcare system, drug failures due to DIKI is a major concern for the pharmaceutical industry, given its frequently reported occurrence in drug discovery and development. Specifically, safety/toxicity-related failures account for 82% of drug project closures,6 and among these, renal injury accounts for 8% and 9% of preclinical and clinical failures, respectively (Fig. 1A). DIKI spans various therapeutic areas, including respiratory/inflammation, cardiovascular/gastrointestinal, and central nervous system (CNS)/pain (Fig. 1B). Notably, of interest to DIKI, only 3.6% of urology drugs and even fewer renal specific drugs progress from phase I to approval in clinical trials.7 This suggests that the kidney has a higher susceptibility to drug-induced injury, as it is exposed to higher concentrations of circulating drugs and/or metabolites, as compared to other organ systems.8,9 There are several factors that contribute to the accumulation of nephrotoxins within the kidney, such as its high vascularity (receiving about 25% of resting cardiac output) and the gradual increase in the concentration of intraluminal nephrotoxins through the reabsorption of the glomerular filtrate.9
Fig. 1.

A) Incidence of preclinical and clinical failures due to renal injury. B) Incidence of project closure due to renal injury by therapy area. See Cook et al. 20146 for original data.
Current approaches for assessing DIKI
DIKI is assessed preclinically in good laboratory practice (GLP) studies as specified in the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines.10 Typically, these studies span 28 days in one rodent and one non-rodent species and may include a recovery period. Within these in vivo studies, DIKI may be detected by altered renal histopathology and/or changes in clinical chemistry endpoints, such as serum creatinine (sCr) or blood urea nitrogen (BUN).11 Similarly, in humans, renal injury is diagnosed using sCr and BUN, historically standard biomarkers for renal function. One advantage of these biomarkers is their functionality, as healthy kidneys should filter them out of the blood into urine. However, they have limitations in detecting human renal injury, as they are influenced by many renal and non-renal factors independent of kidney function such as age, sex, muscle mass, use of supplements, ingestion of cooked meats, medication, and frequency of intense resistance training and exercise.8 Given the inability of diagnostic methods, like sCr and BUN, to detect the initial stages of renal injury and distinguish between DIKI and other types of AKI, many investigators and regulatory agencies are advocating for new approach methods (NAMs) and more sensitive biomarkers. These would aid in eliminating new drug candidates with unfavorable risk–benefit profiles at an earlier stage and with greater accuracy.12 Within the scope of DIKI, several blood/serum and urinary biomarkers have been widely accepted in clinical practice and by governing agencies, (Table 1).
Table 1.
Historic and FDA approved biomarkers for nephrotoxic safety.
| Biomarker | Source/ Medium | Notes | Reference |
|---|---|---|---|
| Albumin (albuminuria) | Urine |
|
8 , 13–17 |
| Beta-2 Microglobulin (B2M) | Urine |
|
8 , 13–18 |
| Blood Urea Nitrogen (BUN) | Blood |
|
8 , 13 , 14 , 19 , 20 |
| Clusterin (CLU) | Urine |
|
8 , 13–18 , 21–23 |
| Cystatin C (CysC) | Urine, Serum |
|
8 , 13–18 , 21 , 22 , 24 |
| Serum Creatinine (sCr) | Serum |
|
8 , 13 , 18 , 19 , 24–26 |
| Kidney Injury Molecule-1 (KIM-1) | Urine |
|
8 , 13–18 , 21 , 22 , 24 , 27 |
| N-acetyl-beta-D-glucosaminidase (NAG) | Urine |
|
13 , 18 , 22 |
| Neutrophil Gelatinase-Associated Lipocalin (NGAL) | Urine, Serum |
|
8 , 13 , 18 , 21 , 22 , 24 |
| Osteopontin (OPN) | Urine |
|
13 , 22 , 28 , 29 |
| Renal Papillary Antigen (RPA-1) | In vivo tissue, Urine |
|
13 , 23 , 30 , 31 |
| Total Protein (Proteinuria, albuminuria) | Urine |
|
8 , 13–17 |
| Trefoil Factor 3 (TFF3) | Urine, Serum |
|
8 , 13–17 , 21 |
Blood/serum origin biomarkers such as sCr and BUN tend to have a long history of research, are highly validated, have been well established for certain disease states, and are less influenced by diet and hydration making them more stable as compared to urinary biomarkers.32 While blood/serum has a number of advantages such as convenience and use in general routine testing, they may only show significant elevations after substantial renal injury has already occurred.8,32 Research has found that urinary biomarkers have the ability to outperform serum markers for certain diseases.32 The advantages of using urinary biomarkers include non-invasive and repeated collection, allowing for long-term monitoring, aiding in early detection. They may also reflect proximal changes in organ function and damage. However, the disadvantages include their limited reflection of different disease areas, the sample variability due to diet or hydration, and the need for further research and validation for clinical application.32
Translating animal results to human risk for DIKI
Establishing the relevancy of toxicity findings in animal studies to humans remains a challenge.12 Animal models, despite being extensively utilized, do not reliably translate to human toxicity nor do they accurately predict adverse events in clinical trials. This discrepancy may be attributed to the limited genetic diversity in laboratory animals.12,33 Aside from GLP studies that are typically conducted in rodent and non-rodent species, the vast majority of experimental work on DIKI is carried out in rodent models, even though the heterogenicity observed in mouse and rodent models does not fully mirror the complexity of what is seen in humans.34 Additionally, researchers may select a single species, gender, and age for experiments,34 which can introduce several limitations when translating animal data to human risk assessments. Typically, single causative agents are studied in animal models, while humans tend to experience numerous factors that lead to injury. Additionally, animal studies are often conducted in young adults, although it is elderly humans who are at the greatest risk for kidney injury.35 Overall, human renal injury is still poorly researched as compared to other organ systems such as the liver.
Currently, there are a number of promising nephrotoxicity biomarkers, such as urinary markers kidney injury molecule-1 (KIM-1), β2-microglobulin (B2M), cystatin C, clusterin, and trefoil factor-3 (TFF-3) (Table 1). These have been accepted as highly sensitive and specific urinary biomarkers by both the US Food and Drug Administration (FDA) and the European Medicines Agency for monitoring DIKI in preclinical studies and on a case-by-case basis in clinical trials.2,8,36 These accepted biomarkers span a broad range of roles in detecting disease progression. For example, KIM-1, NGA, and CLU biomarker are used in early detection and sCr, BUN, Kim-1, and albuminuria37 are used in late detection. Despite demonstrating considerable potential, the correlations between the rise and fall of these biomarkers and the subsequent development of clinically significant nephrotoxicity warrants further research.8
New approach methods (NAMs) in the detection of DIRI
NAMs are methods developed to reduce, refine, or replace animal testing.38 NAMs offer the potential to be faster, less expensive, and more informative than current approaches for toxicological assessment. The FDA’s Center for Drug Evaluation and Research (CDER) defines NAMs broadly, encompassing in vitro, in chemico, and in silico methods.12 While it is unlikely that researchers and regulatory agencies will completely replace whole animal general toxicity studies in drug development,12 the development, validation, and adoption of NAMs provide an opportunity to reduce the number of animals used in testing, refine current methods that still require animals, and replace animal testing whenever possible.38 Currently, there are several new techniques and models, such as microphysiological systems (MPS), quantitative structure activity relationship (QSAR) computer-based models, and in vitro/in silico toxicity prediction tools, being studied and reported in the literature (Table 2). Most recently, there has been a breakthrough in the area of in silico or AI-driven drug discoveries, with companies like Exscientia reporting the first AI-designed drug candidate to enter clinical trials in 2020 and Insilico Medicine reporting a novel AI-designed first-in-class anti-fibrotic drug candidate for a novel target entering Phase I clinical trial in 2021.48 These innovations and novel methods may have utility in improving pre-clinical drug development programs for human risk,12 including the assessment of DIKI.
Table 2.
A list of NAMs currently used in the field of DIKI.
| New Approach Method (NAM) | Notes | Reference |
|---|---|---|
| TIMP2 AND IGFBP7 (NephroCheck®) |
|
8 , 21 , 39–42 |
| Kidney-on-a-Chip |
|
13 , 37 , 43–46 |
| Functional Nephron Number |
|
37 |
| RNA Biomarkers |
|
37 , 47 |
| Stem Cell Therapy |
|
37 |
| QSAR Modeling |
|
4 , 48–50 |
The role of the DIRIL database in NAM research
The development and facilitation of NAMs for the study of DIKI could be expedited and enhanced by a highly annotated list of drugs with DIKI potential. The creation of DIRIL (drug induced renal injury list)51 will provide the opportunity for such an approach since DIRIL is a highly curated collection of single-molecule, oral admission drugs for human use. The aim of DIRIL is to serve as a research tool for the development and refinement of NAMs specific to DIRI. Using a binary (positive versus negative) classification system linked to compound therapeutic application, DIRIL provides an invaluable resource for research and development in nephrotoxicity. It is particularly relevant for enhancing the discovery of new methodologies to assess severity and better classify nephrotoxicity.
Future perspectives
There is a pressing need for new markers to identify and gauge the severity of DIKI at various stages in drug discovery, development, and marketing. Such biomarkers could prove beneficial in dose range toxicology studies, facilitating the transition from discovery to development, or later during GLP toxicology studies (Fig. 2). Alternatively, they could be used to assess DIKI during clinical trials or even much later during the post marketing phases (Phase IV); biomarkers particularly from metabolomics research could be very powerful in the clinical setting and urinary biomarkers of metabolites have been found to provide a valuable perspective into the various physiological and pathological processes.32 Research has also indicated that renal biomarkers hold promise in genomic and mechanistic studies for a better understanding of AKI, which could aid in future drug development.32,52 The potential impact of such biomarkers for DIKI differs by phase; early preclinical detection of DIKI would allow for the redesign or avoidance of nephrotoxic compounds or drugs. The detection of DIKI later during clinical trials could aid in dose escalation and patient selection; and finally, detection during Phase IV would be invaluable in monitoring adverse events in a wider population. Over the last decade the field of DIKI research has been rapidly evolving, while there are a number of challenges and uncertainties there is also an abundance of opportunities.
Fig. 2.

The potential application of biomarkers of DIKI during drug discovery and development. TS: Target selection; LG&LO: Lead generation and lead optimization; CD: Candidate drugs; DIKI: Drug-induced kidney injury; GLP: Good laboratory practice.
Contributor Information
Skylar Connor, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, United States.
Ruth A Roberts, ApconiX Ltd, Alderley Park, Alderley Edge, SK10 4TG, United Kingdom; University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom.
Weida Tong, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, United States.
Author contributions
SC, RR and WT contributed equally to the authoring, reviewing and editing of this manuscript.
Funding
No external funding was received for this work. SC is grateful for the support by an appointment to the Research Participation Program at the National Center for Toxicological Research (NCTR) of the U.S. FDA through the Oak Ridge Institute for Science and Education (ORISE).
Conflict of interest statement. RR is co-founder and co-director of ApconiX, an integrated toxicology and ion channel company that provides expert advice on non-clinical aspects of drug discovery and drug development to academia, industry, and not-for-profit organizations.
The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Disclaimer
This manuscript reflects the views of the authors and does not necessarily reflect those of the Food and Drug Administration. Any mention of commercial products is for clarification only and is not intended as approval, endorsement, or recommendation.
References
- 1. National Center for Health Statistics CfDCaP . Kidney Disease. 2023. Available from: https://www.cdc.gov/nchs/fastats/kidney-disease.htm.
- 2. Al-Naimi MS, Rasheed HA, Hussien NR, Al-Kuraishy HM, Al-Gareeb AI. Nephrotoxicity: role and significance of renal biomarkers in the early detection of acute renal injury. J Adv Pharm Technol Res. 2019:10(3):95–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Cohen A, Ioannidis K, Ehrlich A, Regenbaum S, Cohen M, Ayyash M, Tikva SS, Nahmias Y. Mechanism and reversal of drug-induced nephrotoxicity on a chip. Sci Transl Med. 2021:13(582):eabd6299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Shi Y, Hua Y, Wang B, Zhang R, Li X. In Silico prediction and insights into the structural basis of drug induced nephrotoxicity. Front Pharmacol. 2022:12:1–12. 10.3389/fphar.2021.793332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Awdishu L, Mehta RL. The 6R’s of drug induced nephrotoxicity. BMC Nephrol. 2017:18(1):1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, Pangalos MN. Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework. Nat Rev Drug Discov. 2014:13(6):419–431. [DOI] [PubMed] [Google Scholar]
- 7. Thomas D, Chancellor D, Micklus A, LaFever S, Hay M, Chaudhuri S, Bowden R, Lo A. Clinical development success rates and contributing factors 2011–2020. Washington, DC: Biotechnology Innovation Organization, Informa Pharma Intelligence, Quantitative Life Sciences; 2021. [Google Scholar]
- 8. Griffin BR, Faubel S, Edelstein CL. Biomarkers of drug-induced kidney toxicity. Ther Drug Monit. 2019:41(2):213–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Guo X, Nzerue C. How to prevent, recognize, and treat drug-induced nephrotoxicity. Cleve Clin J Med. 2002:69(4):289–290293-4, 296-7 passim. [DOI] [PubMed] [Google Scholar]
- 10. ICH Guidelines . Available from: https://www.ich.org/page/ich-guidelines.
- 11. Treacy O, Brown NN, Dimeski G. Biochemical evaluation of kidney disease. Transl Androl Urol. 2019:8(Suppl 2):S214–s223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Avila AM, Bebenek I, Bonzo JA, Bourcier T, Davis Bruno KL, Carlson DB, Dubinion J, Elayan I, Harrouk W, Lee SL, et al. An FDA/CDER perspective on nonclinical testing strategies: classical toxicology approaches and new approach methodologies (NAMs). Regul Toxicol Pharmacol. 2020:114:104662. [DOI] [PubMed] [Google Scholar]
- 13. Wilmer MJ, Ng CP, Lanz HL, Vulto P, Suter-Dick L, Masereeuw R. Kidney-on-a-chip technology for drug-induced nephrotoxicity screening. Trends Biotechnol. 2016:34(2):156–170. [DOI] [PubMed] [Google Scholar]
- 14. Blank M, De Felice A, Goodsaid F, Harlow P, Hausner E, Jacobson-Kram D, Taylor W, Thompson A, Throckmorton D, Xiao SA . Review of qualification data for biomarkers of nephrotoxicity submitted by the predictive safety testing consortium, by team BQR; 2009. Silver Spring, MD, USA. Available from: https://www.fda.gov/media/87781/download.
- 15. Dieterle F, Sistare F, Goodsaid F, Papaluca M, Ozer JS, Webb CP, Baer W, Senagore A, Schipper MJ, Vonderscher J. Renal biomarker qualification submission: a dialog between the FDA-EMEA and predictive safety testing consortium. Nat Biotechnol. 2010:28(5):455–462. [DOI] [PubMed] [Google Scholar]
- 16. Sauer JM, Porter AC. Preclinical biomarker qualification. Exp Biol Med (Maywood). 2018:243(3):222–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sauer JM, Porter AC. Qualification of translational safety biomarkers. Exp Biol Med (Maywood). 2021:246(22):2391–2398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Waring WS, Moonie A. Earlier recognition of nephrotoxicity using novel biomarkers of acute kidney injury. Clin Toxicol. 2011:49(8):720–728. [DOI] [PubMed] [Google Scholar]
- 19. Troth SP, Simutis F, Friedman GS, Todd S, Sistare FD. Kidney safety assessment: current practices in drug development. Semin Nephrol. 2019:39(2):120–131. [DOI] [PubMed] [Google Scholar]
- 20. [Internet] M. BUN (Blood Urea Nitrogen) . 2022. 2022 [cited 2024]; Available from: https://medlineplus.gov/lab-tests/bun-blood-urea-nitrogen/.
- 21. Liu X, Guan Y, Xu S, Li Q, Sun Y, Han R, Jiang C. Early predictors of acute kidney injury: a narrative review. Kidney Blood Press Res. 2016:41(5):680–700. [DOI] [PubMed] [Google Scholar]
- 22. CDER/FDA . DDTBMQ000014 FNIH PSTC executive summary final, by program BQ; 2018. Silverspring, MD. Available from: https://www.fda.gov/media/135310/download?attachment.
- 23. CDER/FDA . Review of qualification data for biomarkers of nephrotoxicity submitted by the ILSI-HESI nephrotoxicity working group, by team BQR; 2010. Silverspring, MD. Available from: https://www.fda.gov/media/113664/download.
- 24. Oh D-J. A long journey for acute kidney injury biomarkers. Ren Fail. 2020:42(1):154–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Gowda S, Desai PB, Kulkarni SS, Hull VV, Math AA, Vernekar SN. Markers of renal function tests. N Am J Med Sci. 2010:2(4):170–173. [PMC free article] [PubMed] [Google Scholar]
- 26. Levey AS, Perrone RD, Madias NE. Serum creatinine and renal function. Annu Rev Med. 1988:39(1):465–490. [DOI] [PubMed] [Google Scholar]
- 27. Wen Y, Parikh CR. Current concepts and advances in biomarkers of acute kidney injury. Crit Rev Clin Lab Sci. 2021:58(5):354–368. [DOI] [PubMed] [Google Scholar]
- 28. Sinha SK, Mellody M, Carpio MB, Damoiseaux R, Nicholas SB. Osteopontin as a biomarker in chronic kidney disease. Biomedicines. 2023:11(5):1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Steinbrenner I, Sekula P, Kotsis F, Cube M, Cheng Y, Nadal J, Schmid M, Schneider MP, Krane V, Nauck M, et al. Association of osteopontin with kidney function and kidney failure in chronic kidney disease patients: the GCKD study. Nephrol Dial Transplant. 2022:38(6):1430–1438. [DOI] [PubMed] [Google Scholar]
- 30. Harpur E, Ennulat D, Hoffman D, Betton G, Gautier J-C, Riefke B, Bounous D, Schuster K, Beushausen S, Guffroy M. Biological qualification of biomarkers of chemical-induced renal toxicity in two strains of male rat. Toxicol Sci. 2011:122(2):235–252. [DOI] [PubMed] [Google Scholar]
- 31. Price SA, Davies D, Rowlinson R, Copley CG, Roche A, Falkenberg FW, Riccardi D, Betton GR. Characterization of renal papillary antigen 1 (RPA-1), a biomarker of renal papillary necrosis. Toxicol Pathol. 2010:38(3):346–358. [DOI] [PubMed] [Google Scholar]
- 32. Xue C, Yang B, Fu L, Hou H, Qiang J, Zhou C, Gao Y, Mao Z. Urine biomarkers can outperform serum biomarkers in certain diseases. URINE. 2023:5:57–64. [Google Scholar]
- 33. Olson H, Betton G, Robinson D, Thomas K, Monro A, Kolaja G, Lilly P, Sanders J, Sipes G, Bracken W, et al. Concordance of the toxicity of Pharmaceuticals in Humans and in animals. Regul Toxicol Pharmacol. 2000:32(1):56–67. [DOI] [PubMed] [Google Scholar]
- 34. Becker GJ, Hewitson TD. Animal models of chronic kidney disease: useful but not perfect. Nephrol Dial Transplant. 2013:28(10):2432–2438. [DOI] [PubMed] [Google Scholar]
- 35. Liu KD, Humphreys BD, Endre ZH. The ten barriers for translation of animal data on AKI to the clinical setting. Intensive Care Med. 2017:43(6):898–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Soares S, Souza LC, Cronin MTD, Waaga-Gasser AM, Grossi MF, Franco GR, Tagliati CA. Biomarkers and in vitro strategies for nephrotoxicity and renal disease assessment. J Nephrol Renal Dis. 2020:5(1):35–42. 10.15761/NRD.1000162. [DOI] [Google Scholar]
- 37. Copur S, Tanriover C, Yavuz F, Soler MJ, Ortiz A, Covic A, Kanbay M. Novel strategies in nephrology: what to expect from the future? Clin Kidney J. 2023:16(2):230–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Administration USFD . New approach methods (NAMs); 2022. 2022 [cited 2024]; Silverspring, MD. Available from: https://www.fda.gov/food/toxicology-research/new-approach-methods-nams.
- 39. l’Etoile M. bioMérieux receives FDA clearance for NEPHROCHECK® test on VIDAS®; 2022. Available from: https://www.biomerieux.com/nl/en/journalists/press-releases/biomerieux-receives-fda-clearance-for-nephrocheck--test-on-vidas.html.
- 40. CDRH/FDA . K153165; 2016. Silverspring, MD. Available from: https://www.accessdata.fda.gov/cdrh_docs/pdf15/K153165.pdf.
- 41. CDRH/FDA . K171482; 2017. Silverspring, MD. Available from: https://www.accessdata.fda.gov/cdrh_docs/pdf17/K171482.pdf. .
- 42. CDRH/FDA . K210793; 2022. Silverspring, MD. Available from: https://www.accessdata.fda.gov/cdrh_docs/pdf21/K210793.pdf.
- 43. Ashammakhi N, Wesseling-Perry K, Hasan A, Elkhammas E, Zhang YS. Kidney-on-a-chip: untapped opportunities. Kidney Int. 2018:94(6):1073–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Yu P, Zhu H, Bosholm CC, Beiner D, Duan Z, Shetty AK, Mou SS, Kramer PA, Barroso LF, Liu H, et al. Precision nephrotoxicity testing using 3D in vitro models. Cell & Biosci. 2023:13(1):231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Wu Q, Liu J, Wang X, Feng L, Wu J, Zhu X, Wen W, Gong X. Organ-on-a-chip: recent breakthroughs and future prospects. Biomed Eng Online. 2020:19(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Leung CM, Haan P, Ronaldson-Bouchard K, Kim G-A, Ko J, Rho HS, Chen Z, Habibovic P, Jeon NL, Takayama S, et al. A guide to the organ-on-a-chip. Nat Rev Methods Primers. 2022:2(1):33. [Google Scholar]
- 47. Chorley BN, Ellinger-Ziegelbauer H, Tackett M, Simutis FJ, Harrill AH, McDuffie J, Atabakhsh E, Nassirpour R, Whiteley LO, Léonard J-F, et al. Urinary miRNA biomarkers of drug-induced kidney injury and their site specificity within the nephron. Toxicol Sci. 2021:180(1):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Tropsha A, Isayev O, Varnek A, Schneider G, Cherkasov A. Integrating QSAR modelling and deep learning in drug discovery: the emergence of deep QSAR. Nat Rev Drug Discov. 2023:23(2):141–155. [DOI] [PubMed] [Google Scholar]
- 49. Kwon S, Bae H, Jo J, Yoon S. Comprehensive ensemble in QSAR prediction for drug discovery. BMC Bioinformatics. 2019:20(1):521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Verma J, Khedkar VM, Coutinho EC. 3D-QSAR in drug design-a review. Curr Top Med Chem. 2010:10(1):95–115. [DOI] [PubMed] [Google Scholar]
- 51. Connor S, Li T, Qu Y, Roberts RA, Tong W. Generation of a drug-induced renal injury list to facilitate the development of new approach methodologies for nephrotoxicity. Drug Discov Today. 2024:29(4):103938. [DOI] [PubMed] [Google Scholar]
- 52. Chen J, Chen Y, Olivero A, Chen X. Identification and validation of potential biomarkers and their functions in acute kidney injury. Front Genet. 2020:11:411. [DOI] [PMC free article] [PubMed] [Google Scholar]
