Drug Repurposing Strategies, Challenges and Successes
The commercial promise of this area of ever-increasing interest has yet to be fully realized.
Complete the form below to unlock access to ALL audio articles.
Drug repurposing, also referred to as drug repositioning or reprofiling, is an approach that identifies new clinical applications for approved or investigational drugs outside of the compound’s originally intended uses. This approach commonly results from the serendipitous discovery of off-target effects or newly recognized actions of existing drugs. For example, thalidomide, which was notoriously removed from the market in 1961 after it was found to induce severe congenital disorders, was later repurposed as a treatment for leprosy after clinical case studies suggested potential effectiveness.
Contents
Why is drug repurposing useful?
Traditional drug discovery vs. drug repurposing
On-target vs off-target drug repurposing strategies
Drug repositioning approaches
- In silico screening
- Activity-based screening
Drug repurposing methodologies
Some success stories for repurposed drugs
The COVID-19 pandemic similarly supercharged advancements in drug repurposing, enabled by rapid patient data collection, drug prediction and compound screening. Over 100 unique repurposed drugs were moved into clinical trials for COVID-19 between January 2020 and December 2021, with 4 drugs receiving FDA approval or emergency use authorization and an additional 15 drugs recommended for off-label use by the National Institutes of Health or the Infectious Diseases Society of America.5 In addition to providing a more rapid development timeline in emergency situations, drug repurposing can reduce safety risks and lower costs, and it holds the potential for the discovery of new targets and pathways that can be further exploited to develop novel therapeutics.
The commercial promise of drug repurposing has not been fully realized, primarily due to patentability and market exclusivity challenges. However, the climate may now be changing as there is ever-increasing interest from clinicians, disease-focused foundations and philanthropic organizations that are desperately seeking quick solutions for the patients they represent. There is also great interest from government agencies responsible for military preparedness, biodefense and rapid response to pandemics for the same reasons. Moreover, drug repurposers are now able to use advanced in silico screening enabled by artificial intelligence (AI) and sophisticated tissue- and organ-level in vitro models that more accurately recapitulate human physiology to improve the selection of existing drugs for further pre-clinical testing and, eventually, clinical trials for new indications.
What is drug repurposing?
Many drugs exhibit polypharmacology in that they simultaneously act on multiple targets or disease pathways, regardless of whether they were “targeted” to a specific molecule. Leveraging this phenomenon, drug repurposing reevaluates previously developed compounds for different medical indications and, if successful, implements them in a new clinical context. Drug repurposing can mean repositioning a previously FDA-approved drug or redeveloping an unapproved drug that previously went through some pre-clinical and/or clinical evaluation stages but did not successfully enter the clinic. It also has been used in a more general sense to reposition any chemical, molecule, nutraceutical or even a live biotherapeutic product (probiotic) for a new application for which it was not designed.
Why is drug repurposing useful?
Traditional drug development can be slow, expensive and prone to high failure rates due to poor safety profiles, lack of efficacy or both. Including costs related to out-of-pocket expenses, time and abandoned trials, the average research and development investment for a new chemical entity is more than $2.5 billion per approved product.
It is important to clarify that drug repurposing is often more than simply identifying new uses for a drug that takes advantage of the same mechanism of action defined for the original indication. It is well established that many drugs actually alter the expression or function of multiple molecules and pathways in vivo that extend beyond their single presumed target. It is this multitude of effects that some researchers seek to harness during a drug repurposing program to target previously unknown pathways relevant for novel disease indications. The process of identifying and characterizing these targets can lead to new approaches for targeting particular proteins or other ligands, and these methods can be further exploited for additional indications or for the design of new chemical entities and hence, novel composition of matter intellectual property (IP).
Traditional drug discovery vs. drug repurposing
Drug repurposing can offer advantages over traditional drug discovery in terms of reducing cost, time-to-clinic and risk of failure for a drug product (Table 1). This is facilitated by a shorter pre-clinical stage and a potential reduction in clinical trial requirements if a drug has proven safe in previous Phase I clinical trials. For repurposed drugs, trial requirements may be limited to Phase II and III, which assess the efficacy of the recycled drug for a new indication and ensure that the compound is safe in patients with that particular disease. Although these features make drug repurposing attractive, the IP protections afforded to novel compounds versus repurposed drugs differ, which has significant commercial implications.
Traditional drug discovery frequently results in composition of matter patents that cover the novel chemical entity as well as methods of use patents, which together offer strong protection for the new therapy. In contrast, drug repurposing may only result in a method of use patent that describes the use of the existing drug for a new indication, which offers comparatively weak protection. However, the protection can be stronger and result in a new composition of matter IP in cases where the repurposed drug requires a new formulation or novel delivery mechanism. Commercial protections afforded to repurposed drugs are also limited to three years compared to five years for a new chemical entity. Further exploration of these considerations is covered in the “Remaining challenges in repurposing drugs” section.
Table 1: Comparison of traditional drug discovery and drug repurposing.
Feature | Traditional Drug Discovery | Drug Repurposing |
Cost | >$2.5 billion | |
Time-to-clinic | 3–12 years9 | |
Failure rate | 90–95% | 25–70% |
Pre-clinical investigation | 1. Target validation 2. Compound screen 3. Lead optimization (SAR, drug-like properties, solubility, etc.) | 1. In silico screening 2. Activity-based screens |
Clinical trial requirements | Phases I–III | Phases II and III |
IP protections | Composition of matter and method of use patents | Method of use patents and composition of matter in some cases |
Commercial protections | New chemical entity protection (up to 5 years after FDA approval) | New use/formulation exclusivity (up to 3 years after FDA approval) |
On-target vs off-target drug repurposing strategies
There are different flavors of drug repurposing based on whether on-target and off-target strategies are utilized. The on-target approach is the most straightforward as it involves linking a newly identified disease mechanism to a druggable target and then identifying existing compounds that interact with that target. This can be achieved by first compiling a list of potential targets based on the known biology of a disease or new insights from discovery programs. For diseases with many potential targets, the druggability of these targets can be estimated based on known features, such as the availability of a ligand-binding pocket within a protein. The highest value targets can then be matched with existing drugs originally designed for other applications via their defined molecular targets using databases, such as DrugBank,17 and subsequently evaluated for efficacy using in vitro activity-based assays.
In contrast, the off-target strategy seeks to identify known or unknown side effects of existing drugs that can be repositioned as an on-target effect for a different indication. This approach may utilize in silico and activity-based screening to assemble a list of candidate compounds that are expected to bind to a particular target, modulate a molecular signature, modify a gene network or reverse a phenotype. This approach can be more complex than on-target drug repurposing, but it offers potentially more options to identify an appropriate drug candidate as well as the opportunity to discover novel drug mechanisms, targets and pathways.
Drug repositioning approaches
Drug repositioning approaches can focus on identifying promising drugs for repurposing or on a disease state that lacks adequate treatment options. Drug-focused repurposing may expand the application of an existing drug to new indications based on off-label usage, analysis of abandoned drugs, review of drugs pulled from the market due to safety or efficacy issues or recycling of drugs that have reached the end of patent exclusivity. One commonly used approach uses compound screening, although it can be pursued in multiple ways, which may be broadly broken into two categories: in silico screening and activity-based screening. In many cases, the in silico and activity-based screens complement each other, with the in silico approach providing a means for down-selection resulting in prioritization of candidate compounds for activity-based screening. Conversely, the activity-based screens can also provide additional training data for the in silico approaches, potentially benefiting future development programs, in addition to validating predictions.
In silico screening
In silico drug screening can take many forms depending on the available data and development goals. Broadly, in silico screening translates large-format, high dimensional datasets (e.g., transcriptomic, proteomic or metabolomic profiles) into functional knowledge through a systems-level view of patterns and relationships between biological entities (e.g., genes, proteins, metabolites), often at multiple scales. Traditional in silico approaches have focused on evaluating drug–target interactions by comparing chemical structures of candidate compounds with ones that are known to bind to a target protein or through use of low-throughput docking simulations that require known 3D chemical structures.
Figure 1: A toy subnetwork of interactions between drugs, targets and indications. A. The drug network can be constructed based on known relationships between entities with stronger connections given more weight. In this case, drug A is in pre-clinical development, so its connectivity within the network is sparser compared to the FDA-approved drugs B and C. B. It is possible to infer potential relationships across the network based on indirect connectivity. Drugs A and B have known three-dimensional (3D) chemical structures that are very similar, so it is possible to infer that drug A may also interact with a well-established target of drug B (Potential Relationship A). Although the 3D structural similarity between drugs A and C is unknown, they share a target, and therefore, there is an increased likelihood that aspects of their chemical structure are shared (Potential Relationship B). Finally, because drugs A and B are very structurally similar, drug B could be a potential candidate for drug repurposing for indication b, which does not have an FDA-approved treatment (Potential Relationship C). Credit: Technology Networks.
Activity-based screening
Regardless of the in silico screening method performed, it is essential to carry out activity-based screening to determine the biological activity of a compound in relevant assays and disease models. Optimally, testing is performed across multiple scales to capture the effects of a compound on a disease phenotype, determine its effects on cell viability (as an initial estimate of toxicity), evaluate drug binding to the putative target (if known) and assess efficacy in functionally relevant tissue, organ or whole organism disease models if available. Key considerations when selecting the primary assay include the in silico prediction type (e.g., drug–target binding vs. phenotype modulation), how closely the assay models the disease of interest and the time and/or cost of the assay.
When information about the mechanism and targets for a disease are limited, candidate drugs for repurposing can be assessed by tracking changes in the phenotype during and after treatment using a relevant model system. Because this type of screening is typically the most challenging and time-intensive, it is critical to balance the accuracy of the model system with time and cost constraints. The use of small whole-organism models (e.g. zebrafish or Xenopus laevis (Xenopus)) that capture multi-organ drug responses or human microphysiological systems, such as organoids and microfluidic organ-on-a-chip (organ chip) technology, that more closely recapitulate human physiology and disease states can offer a balance between biological relevance and program limitations.
Drug repurposing methodologies
Multiple methods have been used to identify existing drugs for repurposing as well as to narrow down drug libraries for further in vitro and in vivo testing. One approach uses a disease-oriented methodology that involves matching of diseases with no treatment or ineffective treatments with drugs that exhibit better therapeutic impact. This can be accomplished using computational models that detect similarities between features of disease states and predict that similar drugs can be used. Another approach is therapeutic target-oriented as it looks to identify new targets for a given indication and then match existing drugs to the novel target. This can be accomplished through experimental discovery of new disease mechanisms and computational approaches that uncover druggable targets related to those mechanisms.
While blinded search or experimental screening methods were primarily used to serendipitously identify effective drugs without prior knowledge in the past, in silico drug repositioning can offer greater speed, reduced cost and the potential to identify novel drug mechanisms. Computational methods that may be used to identify and narrow potential drug candidates include the following (Table 2 and Figure 2):
- Target-based
- Structure-based
- Signature-based
- Pathway/network-based
- Knowledge-based
- Clinical data-based
These efforts also can combine in silico drug prediction with retrospective screening in patient databases to evaluate drugs in specific patient populations before further animal or human testing is performed.
Table 2: In silico methods to identify drugs for repurposing.
Method | Definition |
Target-based | The use of protein data, binding pocket information, and known molecular interactions to identify new targets. It can also be combined with network-based approaches to identify new drug–target relationships. |
Structure-based | The use of two dimensional (2D) or 3D chemical structures in combination with known drug activity data to predict novel functions, such as antibiotic activity. This method can also be combined with target-based approaches to assess binding site complementarity between a drug and a target. |
Signature-based | The use of gene or other molecular signatures derived from disease or treatment omics data to identify similarities between diseases or drug mechanisms. Similarities can be used to predict drugs for repurposing. |
Pathway or network-based | Building a disease model consisting of molecular and cellular interactions to develop and confirm hypotheses for drug repurposing projects. These models sometimes include relationships between entities, such as diseases, cellular processes, proteins and small molecules. |
Knowledge-based | The use of automated literature and text mining through natural language processing to search through large bodies of knowledge. Knowledge can be extracted from published papers or reports and patterns identified to link diseases and mechanisms to repurposing candidates. |
Retrospective clinical analysis | The use of electronic health records, clinical trial data, post-marketing surveillance data and insurance records to identify relationships between diseases, drugs and side effects. May be used alone or in combination with other methods. |
Figure 2: In silico methods that can be used alone or in combination to identify and assess the potential properties of repurposed drugs. Credit: Technology Networks.
Some success stories for repurposed drugs
Successes in drug repurposing have traditionally relied on a combination of serendipity and astute clinical and/or pharmacological observations to identify potential off-target impacts of existing drugs and these approaches continue to be essential to drug repurposing. In 2015, the widely used analgesic, aspirin, was approved for new uses in cardiovascular disease and colorectal cancer prevention based on retrospective clinical analyses and pharmacological analyses.
Rare diseases represent an unmet patient need in drug development and are particularly well-suited to drug repurposing for both scientific and commercial reasons. Approximately 95% of the 7,000 rare diseases lack an FDA-approved treatment and many have poorly defined mechanisms of disease pathology;
Similarly, in the case of the rare neurodevelopmental disorder, Rett Syndrome, the in silico tool, network models for causally aware discovery (NeMoCAD), was used to predict drugs that shift a disease transcriptomic signature towards a healthy state.
Drug repurposing approaches have also identified treatments against aggressive cancers for which the standard of care has not changed over multiple decades. By integrating signatures from cancer biopsies and existing drugs, tricyclic antidepressants and related inhibitors of G-protein coupled receptors were predicted to be active against small cell lung cancer (SCLC) and other high-grade neuroendocrine tumors that metastasize early.
There have also been successful efforts to use deep learning models to represent molecular structures in more useful ways and thereby improve their predictive capabilities across large chemical libraries. A significant advance in antibiotics discovery was made recently using a deep neural network model to predict antibiotic activity across molecules that are structurally distinct from traditional antibiotics.37 The model maps molecules into continuous vectors and compares these maps to a database of mapped compounds with known antibiotic properties. Using this approach, an enzyme inhibitor of c-Jun N-terminal kinase, halicin, which was originally developed to treat diabetes, was found to exhibit broad-spectrum antibiotic activity in mice. Halicin is currently undergoing additional preclinical development in the commercial sector in preparation for clinical trials.
This work also has been extended to identify additional antibiotics, especially against challenging pathogens such as Gram-negative and treatment-resistant bacteria.
Human organoids composed of organ-specific stem cells have also been increasingly used for drug repurposing, especially for rapid screening programs during the COVID-19 pandemic. Self-organizing organoids more accurately recapitulate the 3D tissue microenvironment compared to 2D cell culture systems and, thus, can be more predictive of drug function in vivo. Since SARS-CoV-2 primarily infects the respiratory tract, a human lung organoid model was generated and used to screen FDA-approved drugs for repurposing.
In addition to using signature-based, network-based, and retrospective clinical methods for repurposing drugs during the COVID-19 pandemic, numerous drug repositioning efforts used molecular structure-based approaches to target the viral surface spike protein. However, since surface binding sites on the spike protein readily mutate, on-going efforts against COVID-19 and related viruses are focused on identifying highly conserved, internal sites within the spike protein. Molecular dynamics simulations and AI-based blind docking are being used at the Wyss Institute for Biologically Inspired Engineering to identify a potential binding site, predict existing compounds with strong binding affinity and then apply medicinal chemistry to design novel chemical compounds with improved activities based on the molecular structure of the existing compounds.
Medicinal chemistry was also used in combination with a drug repurposing program, which identified an existing compound (SNC80) as a lead candidate to induce slowing of cell, tissue and organ physiology and metabolism to enhance organ preservation.
Collectively, these stories highlight the wide range of approaches and applications that have been pursued for drug repurposing with each instance demonstrating multiple scientific and commercial pathways for therapeutic development. Despite the significant challenges associated with drug repositioning, including misaligned commercial incentives and lack of adequate patent protection for innovations, this approach has contributed to important developments for patients and holds even greater promise for the future.
Remaining challenges in repurposing drugs
Drug repurposing approaches can offer gains in terms of total development cost, time to market and failure rate due to safety concerns; however, repurposed drugs can still fail at later phases of development due to lack of efficacy in clinical populations, similar to novel compounds.
Novel FDA-approved drugs are legally and commercially protected by composition of matter patents and new chemical entity status; however, after these protections expire, multiple manufacturers can make generic versions of the drug. Although the FDA offers three years of market exclusivity for the new use of previously marketed drugs, pharmaceutical companies may have limited ability to recoup the costs of developing the drug for additional indications due to this limited time window and off-label use that devalues the product.
Drug repurposing is also hindered by barriers to accessing data from failed trials and the resulting shelved compounds. Although there is information about most FDA-approved drugs to support repurposing efforts, compounds that do not make it to the clinic often “disappear” when their development is abandoned, with trial data and results left unpublished.
In this article, we explore the evolution of drug repurposing from a completely serendipitous occurrence to a more formalized endeavor that involves advanced computational approaches and bioengineered test beds. Although scientific and commercial challenges remain, the range of potential applications – from rare diseases to infectious disease emergencies to philanthropic efforts – provides hope for patients with few desirable treatment options.
1. Barnhill RL, McDougall AC. Thalidomide: Use and possible mode of action in reactional lepromatous leprosy and in various other conditions. J Am Acad Dermatol. 1982;7(3):317-323. doi:10.1016/S0190-9622(82)70118-5
2. Singhal S, Mehta J, Desikan R, et al. Antitumor activity of Thalidomide in refractory multiple myeloma. N Engl J Med. 1999;341(21):1565-1571. doi:10.1056/NEJM199911183412102
3. Hirsch MS. Azidothymidine. J Infect Dis. 1988;157(3):427-431. doi:10.1093/infdis/157.3.427
4. Fischl MA, Richman DD, Grieco MH, et al. The efficacy of Azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex. N Engl J Med. 1987;317(4):185-191. doi:10.1056/NEJM198707233170401
5. Greenblatt W, Gupta C, Kao J. Drug repurposing during the COVID-19 pandemic: Lessons for expediting drug development and access. Health Aff. 2023;42(3):424-432. doi:10.1377/hlthaff.2022.01083
6. DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: New estimates of R&D costs. J Health Econ. 2016;47:20-33. doi:10.1016/j.jhealeco.2016.01.012
7. Roessler HI, Knoers NVAM, van Haelst MM, van Haaften G. Drug repurposing for rare diseases. Trends Pharmacol Sci. 2021;42(4):255-267. doi:10.1016/j.tips.2021.01.003
8. Nosengo N. Can you teach old drugs new tricks? Nature. 2016;534(7607):314-316. doi:10.1038/534314a
9. Biotechnology Innovation Organization, Informa Pharma Intelligence, QLS Advisors. Clinical development success rates and contributing factors 2011–2020. February 2021. Accessed November 27, 2023. https://www.bio.org/clinical-development-success-rates-and-contributing-factors-2011-2020
10. Deloitte Center for Health Solutions. Seize the digital momentum: Measuring the return from pharmaceutical innovation 2022. January 2023. Accessed January 5, 2024. https://www2.deloitte.com/content/dam/Deloitte/uk/Documents/life-sciences-health-care/deloitte-uk-seize-digital-momentum-rd-roi-2022.pdf
11. Mullard A. Drug repurposing programmes get lift off. Nat Rev Drug Discov. 2012;11(7):505-506. doi:10.1038/nrd3776
12. Mullard A. Bank tests drug development waters. Nat Rev Drug Discov. 2014;13(9):643-644. doi:10.1038/nrd4412
13. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov. 2004;3(8):711-716. doi:10.1038/nrd1470
14. 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;12(7):3049-3062. doi:10.1016/j.apsb.2022.02.002
15. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: Progress, challenges and recommendations. Nat Rev Drug Discov. 2018;18(1):41-58. doi:10.1038/nrd.2018.168
16. Paul SM, Mytelka DS, Dunwiddie CT, et al. How to improve R&D productivity: the pharmaceutical industry’s grand challenge. Nat Rev Drug Discov. 2010;9(3):203-214. doi:10.1038/nrd3078
17. Wishart DS, Feunang YD, Guo AC, et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1):D1074-D1082. doi:10.1093/nar/gkx1037
18. Luo H, Li M, Yang M, Wu FX, Li Y, Wang J. Biomedical data and computational models for drug repositioning: A comprehensive review. Brief Bioinform. 2021;22(2):1604-1619. doi:10.1093/bib/bbz176
19. Hodos RA, Kidd BA, Shameer K, Readhead BP, Dudley JT. In silico methods for drug repurposing and pharmacology. WIREs Syst Biol Med. 2016;8(3):186-210. doi:10.1002/wsbm.1337
20. NIH LINCS Program. lincsproject.org. Updated January 5, 2024. Accessed February 14, 2024. https://lincsproject.org/.
21. Davis AP, Wiegers TC, Johnson RJ, Sciaky D, Wiegers J, Mattingly CJ. Comparative toxicogenomics database (CTD): Update 2023. Nucleic Acids Res. 2023;51(D1):D1257-D1262. doi:10.1093/nar/gkac833
22. Si L, Bai H, Rodas M, et al. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng. Published online 2021. doi:10.1038/s41551-021-00718-9
23. Sperry MM, Novak R, Keshari V, et al. Enhancers of host immune tolerance to bacterial infection discovered using linked computational and experimental approaches. Adv Sci. Published online 2022. doi:10.1101/2021.09.30.462576
24. Dasgupta Q, Jiang A, Wen AM, et al. A human lung alveolus-on-a-chip model of acute radiation-induced lung injury. Nat Commun. 2023;14(1):6506. doi:10.1038/s41467-023-42171-z
25. Bai H, Si L, Jiang A, et al. Mechanical control of innate immune responses against viral infection revealed in a human lung alveolus chip. Nat Commun. 2022;13(1):1928. doi:10.1038/s41467-022-29562-4
26. Ewart L, Apostolou A, Briggs SA, et al. Performance assessment and economic analysis of a human Liver-Chip for predictive toxicology. Commun. Med. 2022;2(1):154. doi:10.1038/s43856-022-00209-1
27. Sperry MM, Oskotsky TT, Marić I, et al. Target-agnostic drug prediction integrated with medical record analysis uncovers differential associations of statins with increased survival in COVID-19 patients. PLoS Comput Biol. 2023;19(5). doi:10.1371/journal.pcbi.1011050
28. The ROADMAP Project. everycure.org/roadmap. Published 2023. Accessed December 5, 2023. https://www.everycure.org/roadmap
29. Mumau M, Irvine A, Ma C, et al. A Translational approach to identifying and targeting TNF signaling in idiopathic multicentric Castleman disease. Blood. 2023;142(Supplement 1):3921-3921. doi:10.1182/blood-2023-188076
30. Novak R, Lin T, Kaushal S, et al. Target-agnostic discovery of Rett syndrome therapeutics by coupling computational network analysis and CRISPR-enabled in vivo disease modeling. bioRxiv. Published online 2022. doi:10.1101/2022.03.20.485056
31. Bonvissuto D. VUMC receives $13 million grant to coordinate multisite clinical trial for Rett syndrome treatment. VUMC Reporter. January 9, 2024.Accessed January 17, 2024. https://news.vumc.org/2024/01/09/vumc-receives-13-million-grant-to-coordinate-multisite-clinical-trial-for-rett-syndrome-treatment/
32. Gordon DE, Jang GM, Bouhaddou M, et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. 2020;583(7816):459-468. doi:10.1038/s41586-020-2286-9
33. Varona JF, Landete P, Lopez-Martin JA, et al. Preclinical and randomized Phase I studies of plitidepsin in adults hospitalized with COVID-19. Life Sci Alliance. 2022;5(4):e202101200. doi:10.26508/lsa.202101200
34. Jahchan NS, Dudley JT, Mazur PK, et al. A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discov. 2013;3(12):1364-1377. doi:10.1158/2159-8290.CD-13-0183
35. Wang J, Byers LA. Teaching an old dog new tricks: Drug repositioning in small cell lung cancer. Cancer Discov. 2013;3(12):1333-1335. doi:10.1158/2159-8290.CD-13-0790
36. Chen B, Ma L, Paik H, et al. Reversal of cancer gene expression correlates with drug efficacy and reveals therapeutic targets. Nat Commun. 2017;8(1):16022. doi:10.1038/ncomms16022
37. Stokes JM, Yang K, Swanson K, et al. A deep learning approach to antibiotic discovery. Cell. 2020;180(4):688-702.e13. doi:10.1016/j.cell.2020.01.021
38. Liu G, Catacutan DB, Rathod K, et al. Deep learning-guided discovery of an antibiotic targeting Acinetobacter baumannii. Nat Chem Biol. 2023;19(11):1342-1350. doi:10.1038/s41589-023-01349-8
39. Wong F, Zheng EJ, Valeri JA, et al. Discovery of a structural class of antibiotics with explainable deep learning. Nature. Published online December 20, 2023. doi:10.1038/s41586-023-06887-8
40. Han Y, Duan X, Yang L, et al. Identification of SARS-CoV-2 inhibitors using lung and colonic organoids. Nature. 2021;589(7841):270-275. doi:10.1038/s41586-020-2901-9
41. Aman J, Duijvelaar E, Botros L, et al. Imatinib in patients with severe COVID-19: A randomised, double-blind, placebo-controlled, clinical trial. Lancet Respir Med. 2021;9(9):957-968. doi:10.1016/S2213-2600(21)00237-X
42. Duijvelaar E, Schippers JR, Smeele PJ, et al. Long-term clinical outcomes of COVID-19 patients treated with imatinib. Lancet Respir Med. 2022;10(4):e34-e35. doi:10.1016/S2213-2600(22)00052-2
43. Drugs for Neglected Diseases initiative. anticov.org. Updated November 17, 2023. Accessed December 18, 2023. https://anticov.org/about-anticov/
44. Halford B. The path to Paxlovid. ACS Cent Sci. 2022;8(4):405-407. doi:10.1021/acscentsci.2c00369
45. Sperry MM, Charrez B, Fotowat H, et al. Identification of pharmacological inducers of a reversible hypometabolic state for whole organ preservation. eLife. 2024;13:RP93796. doi: 10.7554/eLife.93796.1
46. Novac N. Challenges and opportunities of drug repositioning. Trends Pharmacol Sci. 2013;34(5):267-272. doi:10.1016/j.tips.2013.03.004
47. Begley CG, Ashton M, Baell J, et al. Drug repurposing: Misconceptions, challenges, and opportunities for academic researchers. Sci Transl Med. 2021;13(612). doi:10.1126/scitranslmed.abd5524
48. Krishnamurthy N, Grimshaw AA, Axson SA, Choe SH, Miller JE. Drug repurposing: A systematic review on root causes, barriers and facilitators. BMC Health Serv Res. 2022;22(1):970. doi:10.1186/s12913-022-08272-z
49. Loewa A, Feng JJ, Hedtrich S. Human disease models in drug development. Nat. Rev. Bioeng. 2023;1(8):545-559. doi:10.1038/s44222-023-00063-3