Abstract
Targeted drugs and precision medicine have transformed the landscape of cancer therapy and significantly improved patient outcomes in many cases. However, as therapies are becoming more and more tailored to smaller patient populations and acquired resistance is limiting the duration of clinical responses, there is an ever increasing demand for new drugs, which is not easily met considering steadily rising drug attrition rates and development costs. Considering these challenges drug repurposing is an attractive complementary approach to traditional drug discovery that can satisfy some of these needs. This is facilitated by the fact that most targeted drugs, despite their implicit connotation, are not singularly specific, but rather display a wide spectrum of target selectivity. Importantly, some of the unintended drug “off-targets” are known anticancer targets in their own right. Others are becoming recognized as such in the process of elucidating off-target mechanisms that in fact are responsible for a drug’s anticancer activity, thereby revealing potentially new cancer vulnerabilities. Harnessing such beneficial off-target effects can therefore lead to novel and promising precision medicine approaches. Here, we will discuss experimental and computational methods that are employed to specifically develop single target and network-based off-target repurposing strategies, for instance with drug combinations or polypharmacology drugs. By illustrating concrete examples that have led to clinical translation we will furthermore examine the various scientific and non-scientific factors that cumulatively determine the success of these efforts and thus can inform the future development of new and potentially lifesaving off-target based drug repurposing strategies for cancers that constitute important unmet medical needs.
Keywords: Targeted cancer therapy, precision medicine, drug repurposing, off-target, polypharmacology
1. Introduction
In the past two decades groundbreaking advancements in molecular biology and genomics technologies, powerfully manifested in the sequencing of the human genome, have enabled the birth of the concept of personalized or precision medicine1–4, which means to give the right drug to the right patient at the right time, and which revolutionized particularly the field of cancer research and clinical oncology. The development of potent new targeted drugs, such as the therapeutic antibody trastuzumab for HER2-positive breast cancer and the tyrosine kinase inhibitor (TKI) imatinib for BCR-ABL-driven chronic myeloid leukemia (CML)5–8, has not only dramatically prolonged and improved the lives of many cancer patients, but also created profound excitement among scientists in cancer biology and drug discovery across academia, government and pharmaceutical and biotechnology industry. These developments were to large extent fueled by next generation high-throughput DNA and RNA sequencing technologies9–12, which enabled initiatives like The Cancer Genome Atlas (TCGA) and provided a new, genomically founded and mechanistic understanding of the root causes of many cancers and resulted in the identification of an increasing number of novel cancer targets13–16. Arguably, these exciting discoveries could not have had such paradigm-changing impact if not accompanied by similarly important advances in medicinal chemistry and structural biology. These enabled the synthesis of small molecule inhibitors of cancer-related proteins, predominantly tyrosine kinases that in contrast to early inhibitors like staurosporine exhibited not only high potency, but also improved selectivity for the desired targets.
Although these successes created a wave of discoveries of new actionable targets and targeted drugs for various cancers, it quickly became apparent that few of these novel targeted therapies had curative potential. Rather, drug resistance developed in many cases rapidly through a plethora of mechanisms17–19, as has been powerfully illustrated for imatinib in CML as well as the BRAF inhibitor vemurafenib in melanoma20–22. This further increased the demand for new drugs that could overcome resistance to first generation targeted agents. At the same time, these aspects unveiled the limitations and challenges of traditional drug discovery approaches within the pharmaceutical and biotechnology sector23–25, most notably steadily increasing research and development costs, high attrition rates along the course of clinical development, including failure in expensive late-stage clinical trials, thinning drug pipelines and long development times ranging in average between 10 to 15 years from lead discovery to final market approval26. Considering that rapid development of drug resistance, besides of its obvious clinical ramifications, also means a reduced ability of pharmaceutical companies to recover their initial investments into individual drugs and that precision/personalized medicine with its smaller market shares for each drug per definitionem embodies a profoundly different concept from previous and highly lucrative “one drug fits all” blockbuster drug paradigms of the past, this cumulatively put increasing financial pressure on drug discovery companies.
In light of these challenges, the concept of drug repurposing (also known as ‘drug repositioning’) has gained much attention and momentum in recent years25; 27–29. ‘Drug repurposing’ refers to the identification of new therapeutic applications of existing or investigational drugs beyond their initially intended indication. This may even allow ‘drug rescuing’ of compounds that have failed in initial clinical trials. Identifying new therapeutic uses for existing molecules has obvious benefits for patients that may otherwise not have any efficacious therapy options, but it also provides a powerful avenue towards new research tools for interrogating underlying disease biology and not least it provides an opportunity for pharmaceutical industry to increase their earnings from a particular drug. Thus, drug repurposing can complement the undisputedly valuable traditional drug discovery approach while at the same time relieving some of the aforementioned financial pressure on drug discovery companies. This does not mean that drug repurposing approaches do not require additional investments in themselves or are without risks. In fact, drug repurposing approaches are similarly likely to fail in phase II or III trials for lack of efficacy, which is the major reason for drug failure in recent years accounting for ~50–60%30, as traditional drug development approaches for a drug’s original indication23; 31. However, as drug repurposing candidates have gone through significant preclinical testing as well as phase I safety studies, the risk for toxicity-associated failure is usually low. Furthermore, pharmacokinetics and pharmacodynamics profiles, formulations and dosing schedules are at this point well established, which cuts down significantly on further costs and development times24; 25. As a consequence, there is a plethora of successful drug repurposing cases in cancer and many other diseases, which have been comprehensively reviewed elsewhere29; 32–35. Early successes in this field have in addition inspired dedicated efforts to harness this concept in the form of public-private partnerships by the Medical Research Council (MRC) in the United Kingdom and the National Institutes of Health (NIH) in the United States, which founded the National Center for Advancing Translational Sciences (NCATS), with various pharmaceutical companies36; 37.
The majority of repurposing projects can be traced back to a new mechanistic understanding on the biology of a drug’s intended (“canonical”, “cognate”) target or pathway in a different disease. Examples include repurposing of the immunosuppressant mTOR inhibitor rapamycin and its derivative everolimus for renal cancer38; 39, which led to FDA approval, and the still ongoing investigations into the antidiabetic drug metformin as a stimulant of AMPK signaling in various cancers40, aspirin as a COX-2 inhibitor for the prevention of colorectal cancer41, and α-difluoromethylornithine (DFMO) as an inhibitor of ornithine decarboxylase in neuroblastoma42; 43. In all of these cases the pharmacological target protein or pathway is the same in the original and the repurposed indication (Figure 1A). Notably though, small molecule drugs, even clinically approved ones, can have widely varying target selectivity profiles displaying a range of unintended non-canonical targets44. This is the case even for drugs that may fall into the category “targeted therapies”45–49, a term which suggests high precision. Non-canonical or “off-” targets are often regarded as a liability and are usually considered to be responsible for toxic side effects. There are many documented cases where this is indeed true and some undesired off-targets, so-called “anti-targets” like the ion channel protein hERG that is associated with cardiac toxicity, are routinely screened against in early drug development. However, inhibition of many off-targets is biologically neutral and does not cause meaningful effects as even multi-targeted drugs can be relatively safe depending primarily on what particular off-targets are inhibited and in what biological context. Moreover, in some curious cases inhibition of unintended targets may elicit unexpected beneficial effects in a different disease context44, and thus provide a rationale for drug repurposing (Figure 1B). What makes this aspect particularly interesting is the identification of an unanticipated opportunity to elucidate previously hidden biological disease mechanisms. It is these cases of off-target, rather than on-target, based drug repurposing that we would like to discuss in this review, as they, in many cases, spark or at least contribute to these discoveries. While the definitions of “on-target” and “off-target” are straightforward enough (Box 1), a closer look in this context is necessary. For instance, can the receptor tyrosine kinase (RTK) REarranged during Transfection (RET) be considered an off-target of the multi-targeted drug cabozantinib? Similarly, can KIT be considered an off-target of sunitinib? Strictly speaking, both cases clearly fulfill the offered definition as cabozantinib was developed with the intention to inhibit MET and VEGFR-2, not RET, and sunitinib was developed as an inhibitor of VEGFR and PDGFR RTKs, not KIT. However, as most new drug candidates are usually screened against a broader panel of proteins to determine target selectivity, RET and KIT were early on identified to be off-targets of these two drugs50–52. While this is true for many other proteins, too, the fact that both RET and KIT are inhibited potently enough by these drugs and that at the time of the first publication of cabozantinib and sunitinib, both RTKs were already associated with specific diseases, such as thyroid cancer and gastrointestinal stromal tumors (GIST)53; 54, respectively, the therapeutic opportunities of inhibiting these nominal off-targets was immediately recognized and ultimately led to successful repurposing for these tumor types55–58. Thus, there is a temporal element to assigning this designation and RET and KIT are, with some justification, not widely perceived as off-targets of cabozantinib and sunitinib. For the purpose of this review we will therefore exclude cases like these where the off-target repurposing potential is recognized immediately within the first report of a compound, primarily because for this to be the case the underlying biology has to be sufficiently understood a priori. We will instead focus our attention on those drug-off-target relationships, for which repurposing opportunities (and thus the underlying biology) were initially non-obvious and which were discovered only in the course of later studies. Specifically, this means that while we consider ROS1 as a crizotinib off-target that led to successful repurposing in non-small cell lung cancer (NSCLC) with oncogenic ROS1-fusions (see below), the same does not apply any more to the next-generation TKI lorlatinib as, due to the success of crizotinib, this compound was developed with ROS1 as a secondary target in mind59; 60.
Figure 1. On-target and off-target based drug repurposing.

A. Cartoon illustrating the repurposing of drugs (A; B) for different cancers based on their intended targets A and B playing a functional role in cancer. The initial indication may be another cancer or a cancer-unrelated disease. B. Cartoon illustrating the repurposing of a multi-targeted drug (C) for a specific type of cancer based on inhibition of an off-target (G) that plays a functional role in that cancer. The initial indication may be another cancer or a cancer-unrelated disease.
Box 1. Glossary.
Drug repurposing/drug repositioning - the process of finding new therapeutic applications of existing or investigational drugs beyond their original medical indication
Drug promiscuity - binding of a given drug to multiple targets
Cognate target(s) - intended primary or canonical pharmacological target(s). Inhibition causes on-target effects.
Off-target - unintended drug targets; also referred to as “non-canonical” target
Anti-target - an off-target that is not just unintended, but also undesired as its engagement will produce detrimental effects (e.g. toxicity; reduction of efficacy)
Polypharmacology - binding of a given drug to, and functional modulation of, two or more targets that mechanistically contribute to the overall therapeutic effects within a specific indication
In some cases, the biological activity of a drug is not the result of inhibiting only one, but two or more targets, which may be intended targets, off-targets or a combination of both. This phenomenon is referred to as “polypharmacology”, which was initially defined as the binding of a compound to multiple targets61. However, there have been significant advancements in the field since then, suggesting that, while some drugs are more selective than others, essentially every drug binds more than one target and the term is accordingly used currently in different ways. We would therefore like to define it here more narrowly as the binding of a drug to and functional modulation of two or more targets that are mechanistically relevant for the overall biological or therapeutic drug effect in a defined disease (Box 1). Importantly, in this definition “polypharmacology” is not equivalent to “multi-targeted” as it also requires a mechanistic contribution of multiple targets. Traditionally, drug discovery pursues highly selective and potent drugs for one specific target as such high-affinity drugs are considered to maximize intended target inhibition and minimize unwanted off-target binding that may lead to side effects. This “one disease - one target - one drug” model has been successful in the control of cancers that are driven by a strong oncogenic target (Figure 2A). However, inhibition of a single target is often not sufficient to generate optimal therapeutic benefit where the disease displays polygenicity (e.g. cancer, psychiatric diseases) or involves complex biological signaling networks and feedback loops62–67. For instance, RTKs usually share the same canonical downstream signaling pathways and inhibition of one RTK driver can be compensated for by activation of a different RTK. Accordingly, EGFR-mediated bypass signaling confers resistance to ALK or ROS1 inhibition by crizotinib in fusion-kinase driven NSCLC68–70, and MET signaling can counteract EGFR inhibition by gefitinib in EGFR-mutant NSCLC71. Similarly, feedback activation of EGFR in BRAF-mutant colorectal cancer causes primary resistance to the BRAF inhibitor vemurafenib72. In the case of the PI3K–AKT–mTOR pathway, it is noteworthy that inhibition of mTORC1, for instance by everolimus, or knockdown of its downstream target S6K releases a negative feedback loop and paradoxically leads to upregulation of AKT signaling via e.g. mTORC2 activation73–75. Therefore, treating such complex or “smart cancers” requires similarly complex, “smart” therapeutic approaches that target the extended oncogenic signaling network61; 76; 77. This cannot be accomplished by inhibiting a single protein, but cumulative and even partial effects of drugs on multiple targets, including also less potent ones, can exceed that of single target engagement and thus overcome primary and acquired drug resistance64. Multi-targeted approaches, for instance by inhibition of IGF1R, HDACs or AURKA78; 79, can also suppress drug-tolerant persister cells, which survive single target inhibition and from which outright drug-resistant cell populations can arise over time80; 81. Moreover, multi-targeted drugs can be, maybe unintuitively, less toxic than single-targeted drugs as has been observed with non-selective COX-1/COX-2 inhibitors (e.g. naproxen) that compared to selective COX-2 inhibitors (e.g. rofecoxib) are associated with lower cardiovascular risk82; 83. This kind of multi-targeted or network pharmacology may be achieved by developing either drug combinations for different relevant targets or single drugs with polypharmacology mechanisms of action (MoAs) (Figure 2B). Drug combinations consisting of two or more targeted agents can represent an effective strategy in improving the therapeutic response of patients, reducing the likelihood of drug resistance and prolonging remission84–86. Despite the encouraging success of many drug combinations, clinical translation can be limited by various aspects. For instance, it can be difficult to achieve sufficient target tissue concentrations simultaneously for two or more drugs. In addition, establishment of formulations and dosing schedules is more challenging and can negatively impact patient compliance. There may also be additional regulatory, patent or business strategy hurdles that would have to be overcome, particularly if drugs are developed by different pharmaceutical companies and if they are at different stages of clinical development (different patent life, sharing of profits and investments into trials). Finally, one of the most common issues is that of added toxicity, which may be dose-limiting for one or more drugs within a combination and thus limit the efficacy of the entire drug combination. This is especially a concern for drugs with overlapping toxicity profiles. At the same time, considering that the attempt to engage two or more different targets with one drug requires some structural compromises, selectivity may be harder to achieve for a polypharmacology drug compared to a drug designed towards inhibiting a single target. This in turn may increase the probability to inhibit anti-targets and elicit toxicity. However, it is also conceivable that two single-targeted drugs together can be less selective than a single polypharmacology drug. In summary, although rational design of polypharmacology drugs and avoiding increased toxicity is technically challenging and in some cases may actually not be achievable, single polypharmacology drugs may be more advantageous than drug combinations in many cases, as reviewed comprehensively also elsewhere61; 87; 88. Interestingly in the context of repurposing, there are not only some powerful examples where drug polypharmacology has been intentionally designed, but also some recent cases where drugs may be repurposed for specific cancers due to previously unknown and unanticipated polypharmacology mechanisms, which - sometimes exclusively - involved several off-targets and unveiled novel network vulnerabilities of these cancer cells (Box 2).
Figure 2. Single target and network pharmacology strategies for cancer therapy.

A. Monotherapy for targeting a single, strong oncogene driver leading to cancer cell death. B. Network-based targeting strategies for cancers and inhibition of oncogenic pathways driven by multiple oncoproteins of varying strength (arrow width) can be implemented either by combination therapy with more than one drug (upper panel) or by a single drug with polypharmacology mechanism of action against more than one cancer-relevant target (lower panel).
Box 2. Polypharmacology Drugs.
Successful design of polypharmacology drugs [on-target]
Dasatinib was developed as a dual ABL/SRC inhibitor241 as, due to structural similarities between these kinases, SRC inhibitors could potentially inhibit imatinib-resistant BCR-ABL mutants in CML341. In addition, BCR-ABL can activate LYN and other SFKs342, which have been implicated in imatinib resistance in CML and BCR-ABL-positive ALL343; 344. Similar rationales led to development of bosutinib (approved by FDA and EMA) and bafetinib345–347.
Sunitinib is a multikinase inhibitor approved by the FDA and/or EMA for GIST, renal cell carcinoma (RCC) and pancreatic cancer. Independent of targeting specific oncoproteins (e.g. mutant KIT in GIST), sunitinib was designed for dual inhibition of VEGFR and PDGFR RTKs50, which are critical for tumor neoangiogenesis and combined inhibition of which in the tumor microenvironment produces potent antitumor effects in various cancers348. Similarly, sorafenib inhibits multiple kinases, including VEGFRs, PDGFRβ, and exhibits potent antiangiogenic and antitumor effects, although it was primarily designed to target RAF1349; 350. Sorafenib is approved for RCC, hepatocellular carcinoma, and thyroid cancer.
Using iterative chemical synthesis, X-ray crystallography and kinase target profiling, Apsel et al. designed several polypharmacology compounds. PP121 inhibits multiple tyrosine and lipid kinases, such as ABL, mTOR and PI3K, and can override BCR-ABLT315I-mediated imatinib resistance in CML cells351.
Fusing two functional pharmacophores generated bispecific hybrid molecules, some of which are in preclinical or clinical tests, such as the HDAC/EGFR/HER2 inhibitor CUDC-101352; 353 and the HDAC/PI3K/AKT inhibitor CUDC-907354, and the dual HDAC/JAK inhibitor EY3238355. Whereas these compounds combine HDAC and kinase inhibitor scaffolds, the former can also be integrated with inducers of reactive oxygen species356, topoisomerase inhibitors357, or BET inhibitors358. Serendipitous off-target inhibition of BET proteins has also been reported for several kinase inhibitors, such as the clinically advanced JAK2 inhibitor fedratinib359; 360, suggesting a possibility to design polypharmacology drugs with selectivity across the kinase and BET protein families361.
Repurposing opportunities due to novel polypharmacology-based mechanisms [off-target]
The ALK inhibitor ceritinib is approved for crizotinib-resistant, EML4-ALK-positive NSCLC303; 305. Independently, ceritinib was found to display moderate cellular efficacy also in EML4-ALK-negative NSCLC cells. Integrated functional proteomics and network analysis elucidated its polypharmacology in these cells through the off-targets RSK1/2, IGF1R and FAK1. Identification of the downstream effector YB1, which mediates taxol resistance, led to the rational design of synergistic combination with taxanes, detection of a potential mechanistic biomarker and clinical translation131.
Midostaurin, originally characterized as a PKC inhibitor and subsequently found to inhibit many other kinases, such as FLT3 and KIT179; 180, was approved for FLT3-mutated AML and SM. Using a similar approach as for ceritinib (see above), Ctortecka et al. found that midostaurin’s cellular potency in NSCLC cells resulted from polypharmacology involving its off-targets TBK1, PDPK1, and AURKA. This led to identification of midostaurin synergy in these cells with an inhibitor of PLK1, a converging downstream signaling protein of AURKA and TBK1130.
Foretinib and its structural analogue cabozantinib were designed as multi-kinase inhibitors, specifically for MET and VEGFRs, which cooperate to promote tumor angiogenesis51; 293; 362. While cabozantinib received regulatory approval for thyroid cancer and RCC, clinical development of foretinib was subsequently discontinued. However, a recent study in NSCLC observed distinct cellular activities of these almost identical drugs. A systems chemical biology approach identified foretinib’s MoA to involve multiple off-targets, namely AURKB, MEK1/2 and FER, which were also targeted by cabozantinib, but less potently133. Because AURKB is a relevant target in MYC-amplified SCLC363, this led to the identification of foretinib synergy with a more potent AURKB inhibitor in MYC-amplified SCLC.
Having introduced the rationale for drug repurposing and its potential specifically with regard to clinical translation and discovery of new cancer biology arising from repurposing approaches that are based on engaging one or more drug off-targets, we will in the following discuss strategies and methods, both computational and experimental, that are employed to identify new repurposing opportunities. We will furthermore illustrate several cases of off-target based repurposing that have been translated into clinical studies for various cancers or even received regulatory approval and finally review those factors, which have enabled or prevented the clinical success of these endeavors and which therefore will be informative for future off-target based repurposing efforts.
2. Current strategies towards drug repurposing
Before we turn our attention to specific cases of off-target based repurposing that have been translated into clinical studies, it is worthwhile discussing the different strategies developed to identify drug repurposing opportunities, which can be roughly divided into computational and experimental approaches.
Computational Approaches
There is a variety of computational or in silico approaches that have been widely used in the exploration of drug repurposing opportunities, starting from drug target identification, prediction of new indications, to mechanistic investigation in a given indication, which have been excellently reviewed in several recent articles and will be therefore covered here only in brief25; 32; 89. In silico methods exhibit many advantages over experimental approaches with reduced requirements regarding cost, time and labor. These approaches are heavily driven by large quantities of data, such as various omics data sets (e.g. gene expression), the chemical structure space, protein interaction databases, electronic health records, the scientific literature, etc. Powerful examples include the mining of side effect profiles of marketed drugs for potentially shared targets and subsequent target-informed repurposing opportunities90 or harnessing gene expression signature-based Connectivity Map approaches to identify cellular targets and activities of existing compounds in new disease contexts91–93. Other systems biology approaches towards interrogating complex datasets and identifying new drug indications use for instance ‘network pharmacology’ that harnesses drug-gene or drug-protein correlations61; 76; 77; 94–96. The DEMAND hybrid method combines gene expression analysis with network perturbation upon drug treatment to elucidate drug-target interactions97. The similarity ensemble approach (SEA) on the other hand focusses on the chemical structures of small molecule drugs or ligands based on the hypothesis that ligands with similar structures are likely to have similar targets98; 99. Application of this concept has been shown to successfully identify multiple new drug-target interactions, which can lead to rapid repurposing if a new target is associated with a specific disease. In contrast, the Repurposing Drugs in Oncology (ReDO) project is pursuing a literature-based approach to find licensed non-cancer drugs, which may also have utility in cancer based on prior reports of anticancer activity or indirect evidence based on activity against cancer-related pathways100. Finally, over recent years much effort by academia and the pharmaceutical industry has gone into artificial intelligence (AI) techniques with the goal to improve the performance and extend the utility of computational methods and mine the inventory of either existing/known drugs or discontinued drug candidates to discover repurposing opportunities including new indications in cancer (Box 3)101; 102.
Box 3. Artificial intelligence (AI) approaches in drug repurposing.
Al-Ali et al. used support vector machines (SVM), a pattern recognition algorithm, and found some kinase inhibitors to exhibit multi-targeted mechanisms and promote neurite outgrowth suggesting the potential for the treatment of neurodegenerative diseases364. Combining the SVM approach with cell-based compound screening allowed several cancer-selective kinase targets to be identified and subsequently functionally validated in triple-negative breast cancer cell lines365. As the selective target dependencies observed with this method are directly correlated with functional compound screening, the identified targets are inherently actionable thereby potentially providing a short path towards repurposing of hit compounds for the queried disease.
The clinical outcome search space (COSS) platform for drug repositioning is composed of literature search, analysis, and data visualization modules that can generate a multi-level, mechanism-based rationale to support drug-disease associations366; 367. Applying this method, BVA201, an anti-depressant approved in Russia, is being repurposed to treat multiple sclerosis and the antibiotic BVA501 was found to display favorable anticancer activities in glioblastoma, melanoma, and thyroid cancer.
Lim et al. employed an approach termed REMAP, which can perform large-scale off-target predictions using chemical and protein space and their interactome. They proposed by comparison with known cancer drugs that seven cancer-unrelated drugs have repurposing potential for cancer, most of which were supported by experimental evidence368.
Another strategy applies proteome-wide docking of small molecule ligands to available protein X-ray crystal structures in order to identify new drug-target relationships and repurposing opportunities. For instance, application of this approach suggested that the antiviral drug nelfinavir exhibits potential as an antifibrosis compound through inhibition of TGFβRI369. AI in the context of drug repurposing is a rapidly developing, albeit still new, area and it will be interesting to follow its contributions to this field over the next years.
Experimental Approaches
Although targeted approaches have been successful in specific cases, such as imatinib repurposing for gastrointestinal stromal tumors (GIST) (see below), phenotypic screening has been widely recognized as a powerful discovery tool103; 104, which can also drive drug repurposing approaches. Phenotypic screening has been successfully performed with a variety of model systems. In the context of cancer, readouts usually include cancer cell viability, proliferation or death, tumor growth, or more specific phenotypes, which are sometimes preferred due to facilitated mechanistic follow up105, such as using specific cancer-associated isogenic cell line pairs106 or processes like cytokinesis107. Among the most popular cancer models are cancer cell lines108; 109, and several landmark, large-scale cell line screening studies have revealed new drug-gene relationships and repurposing opportunities110–117. Notably, the results of several of these high-throughput screens can be mined through public access portals, such as the Cancer Cell line Encyclopedia (CCLE)110; 113, the Cancer Therapeutics Response Portal (CTRP)111, Genomics of Drug Sensitivity in Cancer Web (GDSC)118, and the NCI Transcriptional Pharmacodynamics Workbench (NCI TPW)119. Although these studies have been highly informative, they screened relatively limited 2D cell culture models. More recent efforts have also utilized more complex 3D models and conditionally reprogrammed cell line cultures derived from patient tumor tissue for small molecule screening120–122, the latter of which also have potential to inform individual therapy decisions123. Along those lines, but economically restrictive for most researchers, Gao et al. recently reported on an HTS screen using a large panel of ~1000 patient-derived tumor xenograft mouse models, which was found to be predictive of clinical trial responses with the screened drugs124. However, organism-based cancer screens have also been described using more accessible, specialized fly or zebrafish models125–129. These different phenotypic screening approaches have been or can be readily used for drug repurposing, particularly when they are further supported by the identification of a potential biomarker derived from drug-gene correlations. However, this has been mostly implemented for on-target-based repurposing, as for instance for PARP1 inhibitors in Ewing’s sarcoma110. To query off-target-based drug effects, it is useful to build some target redundancy into the compound screening library, which will enable distinguishing on- from off-target effects, and subsequently analyze the data for unique, rather than common, drug activities, which has been described in some recent studies130–133. Interestingly, this kind of strategy does not only successfully detect anticancer off-target effects in general, but also identifies those non-canonical anticancer activities, which are the result of complex polypharmacology mechanisms that involve multiple off-targets130; 131; 133. This, in turn, emphasizes the need for subsequent unbiased target identification and deconvolution approaches that are critical for any off-target-based repurposing approach and in particular for polypharmacology-driven mechanisms.
In addition to the computational approaches discussed above, there are many experimental methods that are being used to identify drug targets in an unbiased way, also as primary screening assays for target identification and drug repurposing, which are reviewed in detail elsewhere134–136. While gene expression-based techniques like the Connectivity Map have been shown to be highly useful for target identification, they have limited utility for dissecting off-target effects that are specific for certain cell types or polypharmacology mechanisms. Recombinant protein binding or catalytic enzyme screening assay panels can detect new drug-protein interactions and at the same time provide functional validation of these interactions45; 47–49. However, they are by definition limited to defined subsets of proteins, such as kinases or bromodomain proteins, and therefore may or may not capture the target underlying any observed cellular activity. In contrast, proteomics approaches are essentially unbiased, provide broad coverage of the target space and accordingly have demonstrated high utility to identify drug off-targets. Protein microarrays were first reported in 2000137 and have been used in drug discovery for drug target identification and validation138, but have in recent years been largely replaced in popularity by mass spectrometry (MS)-based chemical proteomics, which also reflect the cellular context of a target, such as post-translational modifications, mutation status and protein expression level139. A chemical proteomics approach, which is widely used for the identification of targets within enzyme families with catalytic activity, is activity-based protein profiling (ABPP)140; 141. Although powerful, this assay, like screening recombinant proteins, is inherently biased towards the detection of defined target classes as ABPP probes selectively label active enzymes, such as phospholipases and kinases142; 143. This also applies in a similar fashion to mixed drug affinity matrix approaches, such as kinobeads144; 145, which combine several broad-spectrum kinase inhibitors to enrich much of the kinome and other ATP-binding proteins. In contrast, the recently developed cellular thermal shift assay (CETSA) can cover several thousand proteins and has been demonstrated to be able to identify drug targets and their protein binding partners in an unbiased way146. Like ABPP and the kinobeads approach, CETSA does not require chemical modification of a compound of interest. However, the biochemical rules governing susceptibility of individual targets to this approach are still not completely understood and not all known drug targets are being captured. Although they require chemical modification, compound-centric approaches, which can be also modified by integrating quantitative MS for determination of dissociation constants147, have been shown to have broad utility across multiple drug and target classes148–151, which makes them well suited for unbiased off-target protein identification of phenotypic screening hits.
3. Clinical translation of off-target-based repurposing in cancer
As mentioned above, there is a large number of studies that report new drug repurposing opportunities, even if limiting this to off-target based repurposing only, and it would exceed the capacity of this article to attempt to cover them all. However, the purpose of this review is not to provide comprehensive coverage of the field, but to illustrate the concept, potential strategies, clinical potential and challenges of off-target based drug repurposing approaches in cancer. To this end, it is most informative to discuss those cases that have been translated into subsequent clinical studies as they more readily allow us to glean some insight into the factors that led to ultimate success, i.e. regulatory approval for the new indication, or in some cases failure to do so, which we expect will both be equally helpful for designing future repurposing projects. Below we are describing some of the most iconic clinical attempts of off-target based drug repurposing (Table 1). The majority of these involve kinase inhibitors, for two specific reasons: 1. Many kinases have been identified to drive various cancers and thus are considered highly relevant cancer targets. 2. Kinase inhibitors are notoriously unselective (although there are exceptions) and therefore make particularly good candidates for off-target based drug repurposing efforts. The path for non-kinase inhibitors, however, frequently stems from reports of unexpected anti-cancer activity in patients being treated for non-cancer related maladies. These cases cover a yet wider range of potential targets and are equally, if not more, interesting. We will therefore discuss examples of both, kinase inhibitors and non-kinase inhibitors, that are being evaluated for off-target repurposing in cancer.
Table 1.
Summary of clinical off-target based repurposing studies
| Drug | Original Target | Original Disease | Off-Target(s) | Repurposed Disease | Clinical Status | Revenue |
|---|---|---|---|---|---|---|
| Crizotinib | MET | Solid Tumors | EML4-ALK | NSCLC | Approved | >$3 bio (2012–18) |
| ROS1 Fusions | NSCLC | Approved | ||||
| Imatinib | BCR-ABL | CML | KIT | GIST | Approved | N/A for GIST alone |
| Midostaurin | PKC | Solid Tumors | FLT3 | FLT3+ AML | Approved | N/A |
| KIT | SM & MCL | Approved | ||||
| Thalidomide | Unknown | Morning Sickness | Immune Modulation (CRBN) | ENL, MM | Approved | ~$4 bio (2005–18) |
| Lenalidomide | MM, MDS (5q), MCL | Approved | >$53 bio (2006–18) | |||
| Pomalidomide | MM | Approved | ~$7 bio (2013–18) | |||
| Erlotinib | EGFR | NSCLC | JAK2 | MDS, AML | Discontinued (Phase II) | - |
| Tozasertib | AURKA-C | Multiple Cancers | BCR-ABLT315I | CML & Ph+ ALL | Discontinued (Phase II) | - |
| Desipramine | SERT, NET | Depression | Various Receptors | SCLC | Discontinued (Phase II) | - |
| Dasatinib | SRC, ABL | CML & Ph+ ALL | DDR2 | NSCLC | Ongoing | - |
| BTK | CLL | Ongoing | - | |||
| Ibrutinib | BTK | MCL, CLL, WMG | EGFR | NSCLC | Ongoing | - |
| ERBB2 | ERBB2+BC & GEC | Ongoing | - | |||
| Axitinib | VEGFRs | RCC | BCR-ABLT315I | CML | Ongoing | - |
| Ponatinib | BCR-ABLT315I | CML | RET | RET+ MTC & NSCLC | Ongoing | - |
| Cabozantinib | MET, VEGFR2 | MTC, RCC, HCC | ROS1 Fusions, NTRK Fusions | NSCLC | Ongoing | - |
| Ceritinib | EML4-ALK | NSCLC | ROS1, | NSCLC | Ongoing | - |
| IGF1R, FAK, RSK1/2 | NSCLC | Ongoing | - | |||
| IGF1R, ACK1 | Melanoma | Ongoing | - | |||
| Itraconazole | Lanosterol-14a-de-methylase | Fungal Infections | SMO | Multiple Cancers, BCC | Ongoing | - |
| Disulfiram | ALDH | Alcoholism | NPL4 | Multiple Cancers | Ongoing | - |
| Chloroquine | Heme | Malaria | PPT1 | Multiple Cancers | Ongoing | - |
| Artemisinin | Reactive Oxygen Species | Malaria | BAD | Multiple Cancers | Ongoing | - |
| Sulfasalazine | Unknown | IBD, RA | SPR | Cancer Pain | Ongoing | - |
BC: breast cancer; BCC: basal cell carcinoma; GEC: gastroesophageal cancer; HCC: hepatocellular carcinoma; IBD: inflammatory bowel disease; MTC: medullary thyroid cancer; RA: rheumatoid arthritis; RCC: renal cell carcinoma; WMG: Waldenstrom’s macroglobulinemia; N/A: not available.
Crizotinib
Crizotinib arguably represents one of the most powerful examples of the transformative potential of off-target based drug repurposing as this TKI was originally developed as a potent inhibitor of the RTK MET152; 153, which is involved in many cancer-related processes including proliferation and metastasis, but ultimately received regulatory approval by the US Food and Drug Administration (FDA) in its capacity as an inhibitor of the fusion kinase EML4-ALK. Although crizotinib’s ability to inhibit NPM-ALK fusions in anaplastic large-cell lymphoma (ALCL) cell lines had been noted, the chromosomal translocation leading to the EML4-ALK fusion oncogene in 3–7% of NSCLC and recognition of its transforming activity were only identified shortly thereafter154. Further studies demonstrated that crizotinib could successfully shut down oncogenic signaling and inhibit the growth of EML4-ALK fusion-positive NSCLC cells155. Subsequent phase I clinical trials focusing on EML4-ALK-rearranged lung cancer patients showed, in addition to a favorable safety profile, tumor shrinkage or stable disease for the majority of patients156; 157. The impressive responses observed in these studies, which were later confirmed in a phase III study158, led to the accelerated approval of crizotinib for EML4-ALK-positive NSCLC in the US in 2011 and Europe in 2012, eventually replacing conventional chemotherapy as first-line therapy.
Inspired by this success, Bergethon et al. showed that in preclinical models, as well as in a NSCLC patient, crizotinib also inhibits the ALK-related RTK ROS1159, which constituted a potent off-target of crizotinib that had not been picked up in the initial reports152; 153. Similar to ALK, ROS1 tyrosine kinase activity is constitutively activated as the result of chromosomal translocation events, which had been described previously to serve as oncogenic drivers in NSCLC155; 160, but in contrast to EML4-ALK involve a variety of different fusion partners and affect a different, and smaller, subpopulation of NSCLC patients161; 162. The finding that crizotinib can potently inhibit the growth of ROS1-rearranged NSCLC cells yielded a different phase I clinical trial specifically for patients with advanced ROS1-fusion positive NSCLC where crizotinib again showed pronounced antitumor activity163, which led to crizotinib obtaining FDA approval as first-line therapy in 2016 also for ROS1-rearranged NSCLC. In fact, inhibition of ROS1-fusions by crizotinib has proven to be highly effective and generally produces more durable responses than most other targeted therapies even when compared to crizotinib treatment of ALK-rearranged NSCLC, which has been in the meantime replaced as first-line therapy by yet more potent second-generation ALK inhibitors, such as alectinib, initially developed to overcome ALK point mutation-mediated crizotinib resistance164; 165. However, the twofold success of crizotinib repurposing in NSCLC, which was based on the ALK- and ROS1-fusion kinases as off-targets, has made a major clinical impact and has sparked a vibrant research field on targeting oncogenic fusion kinases not just in lung cancer, but in a broad range of tumor types162.
Imatinib
The development of the ABL TKI imatinib and its groundbreaking success in BCR-ABL-driven chronic myeloid leukemia (CML) has revolutionized the field of cancer research and is widely regarded as the paradigm for targeted therapy7; 166; 167. Not surprisingly, therefore, the enthusiasm generated by imatinib also rapidly kindled many repurposing studies for this drug. While imatinib was first rationally developed to inhibit BCR-ABL and PDGFR6; 168, it was subsequently found to be also a similarly potent inhibitor of KIT, which shares high homology with PDGFR169 and for which gain-of-function mutations causing malignant transformation had shortly before been described in gastrointestinal stromal tumors (GIST)54. After observing sustained objective responses in more than half of patients with advanced unresectable or metastatic GIST170, imatinib was further tested in GIST patients with activating mutations of KIT or PDGFRA, which can also show sensitivity to imatinib171, where it also produced significant clinical benefit172. These studies led to regulatory approval of imatinib for the treatment of GIST in 2002. Interestingly, although the more potent second-generation BCR-ABL inhibitor nilotinib was found to be superior to imatinib in patients with CML173, a randomized phase III trial in unresectable or metastatic GIST did not recommend use of nilotinib over imatinib as first-line treatment for GIST174, illustrating that just because a drug is superior over another for inhibiting their cognate target, the same is not always true for therapeutically relevant off-targets. In addition, imatinib in combination with the anti-estrogen therapy letrozole for hormone receptor-positive metastatic breast cancer expressing KIT or PDGFRβ was well tolerated, but appeared to have limited efficacy175. In contrast, clinical studies of imatinib in metastatic melanoma with KIT aberrations showed significant benefit176; 177, although this has been suggested to be limited to KIT mutations rather than cases with gene amplification only178. Thus, while repurposing of imatinib for cancer indications driven by its off-target KIT have been highly successful, particularly for GIST, it is also apparent that understanding the biological context of the respective off-target and its relative importance for driving specific cancers is critical and cannot be implied based on what is known in other tumor types.
Midostaurin
Midostaurin (PKC412) was originally identified as an inhibitor of protein kinase C (PKC)179 and subsequently shown to inhibit other kinases including VEGFR2, PDGFR and KIT180. Interestingly, development of midostaurin as a PKC or VEGFR2 inhibitor in various cancer types ultimately failed due to insufficient efficacy181. However, having screened a panel of small molecules, Weisberg et al. demonstrated the efficacy of midostaurin in inhibiting mutant FLT3 receptor as a novel off-target in leukemia cells182. This was based on the identification of activating mutations in the RTK FLT3, particularly internal tandem duplications (ITD) in the juxtamembrane domain, in approximately 30% of acute myeloid leukemia (AML) patients183. This finding was followed by clinical observations showing efficacy of midostaurin in AML patients with activating FLT3 mutations, although it was recognized that combination with conventional chemotherapy may be required184. Subsequently, midostaurin was confirmed to be safe and efficacious in clinical trials and in 2017 received regulatory approval for use in FLT3-mutant AML181; 185; 186.
As described above, another off-target of midostaurin is KIT. D816V mutations of KIT, which are resistant to imatinib187, have been found to play a crucial role in the majority of systemic mastocytosis (SM) cases, a rare hematologic neoplasm that can develop into mast cell leukemia (MCL). Midostaurin was shown to inhibit KIT D816V and block growth of KITD816V-transformed cells at nanomolar concentrations188. This led to a multicenter phase II study of midostaurin in aggressive SM (ASM)/MCL patients in which midostaurin was safe and elicited clinically relevant and durable responses189; 190. Following these studies, midostaurin was approved also for advanced SM and MCL181; 191. It is interesting to note that, similar to crizotinib, midostaurin was ultimately approved for treating malignancies that were driven by off-targets, but not actually for diseases driven by its cognate target PKC, which powerfully illustrates the potential of investigating drug off-target effects. In contrast to crizotinib and imatinib, however, midostaurin’s journey took almost 20 years, which in part is the consequence of a complicated international study in AML. Unfortunately, this also limited the ability and interest of the midostaurin developing pharma company, in this case Novartis, to pursue further repurposing opportunities with midostaurin, such as following up the unexpected observation that midostaurin can potently inhibit the EGFR T790M gatekeeper mutant, but not wild-type (wt) EGFR48; 192; 193, which constituted a high unmet medical need in NSCLC at that time.
Thalidomide
Thalidomide is the infamous founding member of a (re)emerging class of drugs, the history of which is dating back to 1956, when it was introduced as a sedative and hypnotic that also alleviates morning sickness in pregnant women194; 195. However, just as imatinib is the paradigm for the success of targeted therapies, thalidomide can be considered the paradigm for severe drug toxicity as it exhibits dramatic teratogenic properties and led to thousands of infants that were born with limb deformations196; 197, such as phocomelia, which later resulted in thalidomide withdrawal from the market198. Strictly speaking, the thalidomide case does not qualify as off-target-based repurposing considering that the pharmacological target was unknown for the longest time. However, its initial indication was based on thalidomide structural and medicinal similarity with barbiturates, which implied a similar target. After thalidomide had made a serendipitous comeback in 1965 as a treatment for erythema nodosum leprosum, an immunological complication of leprosy199, for which it gained FDA approval in 1998, it was also recognized that thalidomide displays potent immunomodulatory properties (hence the term ImmunoModulatory Drug or “IMiD”) by e.g. inhibiting TNFα synthesis200; 201. Subsequently, a possible association of thalidomide with malformations due to inhibition of angiogenesis was identified, which was likely independent of TNFα202. As multiple myeloma (MM) features enhanced neovascularization in the bone marrow, thalidomide was repurposed for the treatment of MM203, for which it showed remarkable efficacy as single agent and in combination with dexamethasone and eventually obtained regulatory approval. These successes subsequently led to the development of more potent and stable IMiDs, namely lenalidomide and pomalidomide, which can enhance the cytokine release and anti-myeloma activity of thalidomide204–208 and are also effective in mantle cell lymphoma (MCL) and myelodysplastic syndrome (MDS)209; 210. While these analogues were being developed in the clinics other studies were progressing on identifying the MoA of thalidomide and the IMiDs. Ito et al. proposed that thalidomide causes its teratogenic effects by binding to the E3 ubiquitin ligase cereblon (CRBN) in complex with damaged DNA binding protein 1 (DDB1) and Cul4A thereby inhibiting its ubiquitin ligase activity211. Further refinement of this mechanism showed that IMiD binding to CRBN leads to recruitment, ubiquitination and selective degradation of the lymphoid transcription factors IKZF1 and IKFZ3212–214. Based on this discovery, Winter et al. designed novel thalidomide-based proteolysis-targeting chimeric molecules (PROTACs) that efficiently and selectively led to degradation of the transcriptional coactivator BRD4 or the chaperone FKBP12215. This provided a powerful demonstration of the broad applicability and potential of the PROTAC approach216 for the design of novel drug candidates and unique chemical probes and invigorated academia and pharmaceutical industry alike as potentially any protein of interest, which could be considered engineered indirect thalidomide off-targets, could be targeted for degradation. Even though this is a developing field and many design rules have to be still established, there is a lot of momentum and the first PROTACs are entering clinical trials already, a truly remarkable development considering thalidomide’s tragic past.
Erlotinib
Erlotinib has been developed as a specific TKI for the epidermal growth factor receptor (EGFR)217. EGFR shows TKI-sensitive oncogenic gain-of-function mutations within the tyrosine kinase domain in about 10–15% of NSCLC patients218; 219, which represents erlotinib’s primary clinical indication and which transformed the field of lung cancer therapy220. Interestingly, it has been observed that erlotinib is also an inhibitor of JAK2 V617F, the dominant mutation in polycythemia vera (PV), and inhibited PV cell growth221. In addition, Boehrer et al. suggested that erlotinib exhibits antineoplastic off-target effects in AML and MDS via JAK2222, although other mechanisms via inhibition of other targets have been proposed as well223. Following these findings, there have been multiple efforts to repurpose erlotinib for targeting of JAK2 in the clinics. Although erlotinib appeared to induce a significant number of responses in higher-risk MDS/AML having failed azacitidine treatment in a phase I/II study224, Komrokji et al. found despite general tolerability only modest single-agent activity of erlotinib in a phase II study in MDS225. Similarly, erlotinib as a single agent was found to have only limited clinical efficacy in patients with relapsed/refractory AML in a phase II clinical trial226. Collectively, these studies suggest that while JAK2 may constitute in general an interesting off-target of erlotinib, it may not have the required potency for producing clinically meaningful effects and/or that JAK2 may not be a strong enough driver for AML, which is in line with reports that also the potent JAK2 inhibitor ruxolitinib may only exhibit clinical efficacy in patients who developed AML from myeloproliferative neoplasms (MPN)227. Accordingly, there are no currently ongoing erlotinib off-target based clinical trials in AML or MDS.
Tozasertib
Tozasertib (VX680) is a potent and selective small-molecule inhibitor of Aurora kinases and considered to possess potential to be used in the treatment of multiple human malignancies, including colorectal cancer, lymphoma and acute leukemias228. By screening clinically relevant mutants against a panel of small molecules and X-ray crystallography, tozasertib was subsequently shown to also bind and have inhibitory activity against BCR-ABL bearing the T315I “gatekeeper” mutation229; 230. This mutant confers broad cross-resistance against the first and second-generation BCR-ABL inhibitors imatinib, dasatinib and nilotinib and represents the most pressing remaining challenge in CML therapy231. Early case reports showed that tozasertib is active in patients with CML and ALL with the BCR-ABL T315I mutation232; 233. However, although tozasertib also showed significant activity in patients with CML or ALL expressing BCR–ABL T315I in a phase I/II trial234, Seymour et al. afterwards observed minimal efficacy of tozasertib and only at higher, intolerable doses235. In addition, there have been reports of cardiac toxicity, such as QTc interval prolongation, which together led to discontinuation of further clinical development of tozasertib for BCR-ABL T315I-mutant leukemias236.
Desipramine
Desipramine is a member of the class of tricyclic antidepressants (TCAs). The TCAs act mainly as serotonin and norepinephrine reuptake inhibitors (SNRIs) by blocking the serotonin transporter (SERT) and the norepinephrine transporter (NET) resulting in enhanced neurotransmission237. TCAs like imipramine and desipramine have been observed in several studies to show anticancer effects for instance in glioma and colorectal cancer cells238; 239, although the underlying mechanisms were not well understood in these studies. Later, Jahchan et al. utilized an innovative bioinformatic drug repurposing approach to compare disease and drug-induced gene expression signatures in small cell lung cancer (SCLC)240. This method identified TCAs to elicit gene expression changes that were anticorrelated to SCLC suggesting TCAs might be efficient drugs to treat this cancer. Indeed, they found TCAs to exhibit in vitro and in vivo anticancer activity in SCLC, which they attributed to a polypharmacology mechanism through inhibition of the cognate targets SERT and NET, but also off-targets like certain histamine, adrenergic, acetylcholine and serotonin receptors240. These observations led to a subsequent phase IIa trial of desipramine in SCLC and other high-grade neuroendocrine tumors. Unfortunately, this trial had to be terminated as SCLC patients were not able to tolerate clinically relevant doses of desipramine and lower drug doses yielded no efficacy, which illustrates that drug doses that are generally safe for the original indication may be too toxic in the context of specific other diseases.
Whereas for the examples discussed above clinical studies have been completed, which allow us to evaluate the success and, in some cases, the encountered challenges, multiple off-target based repurposing attempts for cancer have been translated into clinical trials, which are still ongoing and for which the final outcome is therefore not yet known. However, as it is already a major achievement to launch a clinical trial based on one’s preclinical research, reviewing the trajectory of these projects can allow a better understanding of what aspects facilitated clinical translation. Below, we will therefore discuss several of these cases.
Dasatinib
Dasatinib, a multi-kinase inhibitor that was originally designed as a dual SRC/ABL inhibitor241, received accelerated FDA approval in 2006 as second-line therapy for adults in all phases of CML or with Philadelphia-chromosome-positive (Ph+)-acute lymphoblastic leukemia (ALL). It has since been approved for first-line treatment in both adults and children, thus exemplifying its therapeutic safety when taken at lower doses that are sufficient to inhibit BCR-ABL and SRC-family kinases (SFKs). Having shortly thereafter found activity of dasatinib in NSCLC cells due to a combined inhibition of SFKs and its weak off-target EGFR, Haura et al. began a clinical trial to test the combination of dasatinib and the more potent EGFR inhibitor erlotinib (Figure 3A)242–244. Two patients from this trial had partial responses; one with an EGFR mutation, which can explain the response to erlotinib-based therapy, the other without. This latter patient was instead found to carry a novel oncogenic discoidin domain receptor 2 (DDR2) kinase domain mutation245. Importantly, DDR2 and its closely related kinase DDR1 had previously been identified as potent dasatinib off-targets144; 246; 247. However, this was the first study to identify and quantify the prevalence of DDR2 mutations in lung squamous cell cancer (LUSQ), where they represent approximately 4% of all cases245. In light of these discoveries, dasatinib monotherapy clinical trials in LUSQ and other DDR2-mutant cancers were initiated. Unfortunately, at the dose used (140 mg vs 100 mg for CML), limited efficacy was achieved before toxicities, such as pleural effusions and immunosuppression leading to life-threatening infections led to trial termination. These findings were well aligned with inhibition of SFKs and dasatinib’s potent off-target Bruton’s tyrosine kinase (BTK)248; 249. It has now been shown that several compensatory signals are up-regulated upon dasatinib treatment in LUSQ cells, which limit the efficacy of targeting DDR2250. In particular, the RTKs EGFR and MET are activated suggesting that inhibition of DDR2 alone is not going to be an effective strategy. Rather, a combination of dasatinib, or a yet more potent DDR2 inhibitor251; 252, and a potent MET or EGFR inhibitor (as given to the first patient with DDR2-mutant LUSQ) may be needed for successful DDR2-based repurposing of dasatinib250. This would be similar to what has been described in BRAF-mutant colorectal cancer where EGFR signaling was found to mediate primary vemurafenib resistance72. However, given that dasatinib is included in the active MATCH Screening Trial, where patients are matched to various targeted inhibitors based on their genetic profiles, e.g. DDR2 mutations, efforts to determine dasatinib efficacy are still somewhat ongoing.
Figure 3. Off-target based drug repurposing initiatives for dasatinib and ibrutinib in various cancers.

A. Repurposing of the dual SRC/ABL inhibitor, developed to treat CML, for various B cell malignancies and NSCLC due to its ability to also inhibit BTK and EGFR (weak), respectively. Combination therapy with the potent EGFR TKI erlotinib led to the identification of mutant DDR2 as a novel cancer driver in LUSQ. B. Repurposing of the irreversible BTK inhibitor ibrutinib, developed to treat different B cell malignancies, for breast cancer (with trastuzumab) and EGFR-mutant NSCLC due to its ability to also inhibit HER2 and EGFR, respectively. * indicates oncogenic mutation; ** indicates gene amplification; light bulb indicates discovery of new drug-protein interaction.
In some cases, repurposing is based on an off-target that was originally reported to cause an unwanted side effect. Interestingly, BTK, which was found to be involved in the immunosuppressive side effects of dasatinib247–249, has been implicated as a cancer driver in, for example, diffuse large B-cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL). In fact, the first-in-class covalent BTK inhibitor, ibrutinib (PCI-32765), was also in its infancy at this time253–255. Interestingly, dasatinib has been shown to work in both ibrutinib-sensitive and - resistant DLBCL and CLL cells256–259. However, some reports suggest that activity in these cells is not only due to BTK inhibition, but that also SFKs play a role. For instance, in DLBCL cells carrying the BTK-C481S ibrutinib-resistant mutation, the autophosphorylation site (Y233) of BTK-C481S remains phosphorylated upon dasatinib treatment, while downstream FYN inhibition mediates dasatinib activity257. This suggests a polypharmacology effect in wild-type BTK cells, where inhibition of both SFKs and BTK contributes to the therapeutic activity of dasatinib. A clinical trial using dasatinib in CLL where SFKs, BTK and the related TEC kinase are being evaluated as biomarkers for response has been completed, but results have not yet been reported. These observations with dasatinib are powerful examples of how off-targets can inform new research directions, identify responder patients, lead to the elucidation of new cancer biology mechanisms and even turn initially toxic off-target effects into therapeutic approaches for other malignancies in a similar way as seen with thalidomide. However, they also illustrate that such repurposing strategies, just as much as conventional targeting approaches, may be limited due to adaptive signaling that causes resistance to single-agent therapies.
Ibrutinib
Ibrutinib represents an exciting “rags-to-riches” development story. Originally designed as a tool compound to study BTK biology and to develop potent, non-covalent, BTK inhibitors, ibrutinib was purchased from Celera Genomics in 2006 by Pharmacyclics for $3 million (cash and stock). Just 5 years later, in 2011, Johnson and Johnson paid Pharmacyclics almost $1 billion (upfront and milestones) to co-develop ibrutinib for CLL and non-Hodgkin’s lymphoma260. Having received FDA approval for mantle cell lymphoma (2013), CLL (2014) and Waldenstrom’s macroglobulinemia (2015), AbbVie outbid Johnson and Johnson to buy Pharmacyclics for $21 billion and its success as a BTK inhibitor continues. In addition, inhibition of ibrutinib’s off-targets EGFR and ERBB2/HER2 was indicated from kinase assays in the original report, but it wasn’t until 2014 that ibrutinib was recognized as a potential therapeutic option for EGFR-mutant NSCLC or ERBB2-positive breast cancer (Figure 3B)254; 261; 262. In terms of EGFR-driven cancer, it is uncertain, if ibrutinib can compete with the potent and mutant-selective EGFR inhibitor osimertinib (AZD9291), which in 2018 received FDA approval for first-line treatment of EGFR-mutant NSCLC; however, there remains at least one clinical trial for ibrutinib in previously treated EGFR-mutant NSCLC, which is still active263–266. It could be that combining ibrutinib with other compounds, such as the gold complex auranofin, a thioredoxin reductase 1 inhibitor that inhibits AKT/mTOR signaling in lung cancer cells, will be necessary for effective repurposing in EGFR-mutant NSCLC267. Interestingly, ibrutinib’s ability to inhibit myeloid-derived suppressor cell and mast cell function via BTK and the related interleukin-2-inducible T-cell kinase (ITK) could support the anti-tumorigenic potential of ibrutinib as an ERBB2/HER2 inhibitor268–270, which would represent an intriguing polypharmacology mechanism involving targets in tumor microenvironment cells as well as cancer cells, and clinical trials are currently recruiting for ERBB2-driven gastroesophageal cancer and metastatic breast cancer. Finally, using ibrutinib as an ERBB4 inhibitor in ERBB4-expressing cancers may be possible and ERBB4 is being monitored as one of several possible biomarkers in a phase II study for ibrutinib in refractory metastatic cutaneous melanoma271; 272. While the future looks bright for ibrutinib, paying attention to protocols for handling side-effects coming from years of use in hematologic malignancies will certainly benefit its further off-target repurposing potential273.
Axitinib
Axitinib, an angiogenesis inhibitor whose initial targets were reported to be the VEGFR kinases, PDGFR-β, KIT and FGFR-1, was approved in 2012 for advanced renal cell carcinoma274; 275. While screening 252 clinically relevant drugs for BCR-ABL T315I-gatekeeper inhibitors, Pemovska et al. noticed that axitinib, as opposed to other VEGFR inhibitors, showed strong potency against the assayed cells, suggesting an off-target effect276. Indeed, they found that while axitinib binds the T315I mutant in a range similar to VEGFR2, and at a much lower concentration than parental BCR-ABL, it binds in a very different conformation to ABL than VEGFR2 and differently than other ABL inhibitors, including ponatinib, the only approved BCR-ABLT315I inhibitor. This suggested a different wt/mutant-binding profile between the two drugs276. It has since been shown that axitinib has a very narrow BCR-ABL mutant binding profile and often at concentrations that are clinically unachievable277. However, it could be an option for patients who cannot tolerate ponatinib treatment, which is important given the propensity for patients to develop resistance via this important mechanism. Furthermore, a clinical phase I/II trial in combination with bosutinib for CML patients is currently ongoing, which is particularly interesting as a previous study had found that bosutinib in combination with the Aurora kinase and BCR-ABLT315I inhibitor danusertib exhibited strong synergy in BCR-ABLT315I-positive CML cells due to additional effects of bosutinib on its off-target MEK1/2278.
Ponatinib
Imatinib, as has been mentioned, has revolutionized personalized medicine and specifically CML therapy. Still, disease progression via BCR-ABL mutation rendering the fusion kinase resistant to inhibition remains a major problem even with the development of second-line BCR-ABL inhibitors such as nilotinib and dasatinib. Ponatinib was therefore developed to inhibit a panel of BCR-ABL mutations, but in particular the highly resistant T315I-gatekeeper mutation279; 280. From the beginning, it was understood that ponatinib was a multi-kinase inhibitor with activity also against the therapeutically relevant VEGFR, FGFR, PDGFR and FLT3 kinases279–283. Given that ponatinib was designed specifically to target gatekeeper mutations, which are commonly observed upon initial drug therapy in many kinases, such as EGFR, NPM-ALK and EML4-ALK164; 284–286, de Falco et al. and Mologni et al. both tested and found that ponatinib was a potent inhibitor of the RET gatekeeper V804M and V804L mutants as well as wt RET287; 288. While sporadically found in lung and colon cancer, RET-fusions and germline mutations are the main oncogenic kinases in different thyroid cancer subtypes289 and ponatinib is in phase II clinical trials for multiple tumors, including RET mutation-positive thyroid cancer. Results from a recently completed phase II trial of ponatinib in RET-fusion-positive NSCLC, which was also found to be sensitive to ponatinib290, are not available yet. A recent review summarizes nicely the broad application of ponatinib based on both on- and off-target driven diseases291.
Cabozantinib
Cabozantinib is approved for treating thyroid cancer, renal cell carcinoma, and hepatocellular carcinoma. It has been designed as an inhibitor of mainly MET and VEGFR2, but also RET, AXL, KIT, FLT3, and TIE2 among others were originally recognized as important therapeutic targets of cabozantinib51; 52; 58. Interestingly, VEGFR2 and MET inhibition have been suggested to work together to elicit cabozantinib’s therapeutic effect, as tumors have been shown to compensate for VEGFR inhibition through MET signaling, making cabozantinib an excellent example of effective drug polypharmacology51; 52; 292; 293. More recently, cabozantinib has been recognized also for its ability to inhibit ROS1- and NTRK-fusion kinases that have become resistant to the first-line inhibitor crizotinib or larotrectinib, respectively51; 52; 292–297. While ROS1-fusions are present in 1–2% of NSCLC159, NTRK-fusions have been identified at low rates in several cancers298; 299. However, some cancers, such as mammary analogue secretory carcinomas are 100% positive for NTRK-fusions, thus illustrating the importance of inhibiting these kinases162; 300. Cabozantinib crosses the blood-brain barrier and case studies have shown it to be effective in patients with ROS1-positive, crizotinib- (and ceritinib-) resistant NSCLC primary tumors and brain metastases301; 302. At least two clinical trials are currently ongoing for cabozantinib in ROS1- or NTRK-fusion positive NSCLC and based on these off-targets cabozantinib is one of few promising contenders for second-line therapy in these tumors.
Ceritinib
Ceritinib belongs to the growing family of FDA-approved ALK inhibitors that includes crizotinib, alectinib, brigatinib and lorlatinib and was the first ALK inhibitor approved for crizotinib-resistant EML4-ALK-positive NSCLC303–305. As with cabozantinib, ceritinib has also been found to inhibit ROS1 as a major off-target60; 306. Accordingly, ceritinib is in several clinical trials for ROS1-fusion-positive NSCLC and first results suggest potent clinical efficacy of ceritinib also for this indication307.
In addition to ROS1, other off-targets of ceritinib have been identified. For instance, IGF1R, reported early on305, has been shown to contribute to the overall therapeutic effect of ceritinib in EML4-ALK-positive NSCLC cells308. Different studies also found that IGF1R inhibition is responsible for ceritinib activity in rhabdomyosarcoma and hepatocellular carcinoma309; 310. Through phenotypic screening followed by an integrated functional proteomics approach, another study found that inhibition of IGF1R together with the additional off-targets FAK and RSK1/2 contributes to a more complex polypharmacology mechanism of ceritinib leading to its activity in EML4-ALK-negative NSCLC cells131. It was also observed that these targets modulate a downstream pathway involved in paclitaxel resistance and accordingly that combination with taxanes can lead to pronounced synergy. This was particularly the case in cells with high FAK autophosphorylation, which led to a phase I/II clinical trial for EML4-ALK-negative, EGFR-wild-type NSCLC. Finally, IGF1R and/or ACK1 appear to contribute to ceritinib activity in BRAF/NRAS-wild-type melanoma, in which ceritinib in combination with the MEK inhibitor trametinib showed pronounced in vitro and in vivo anti-tumor activity311. A phase II clinical trial of single-agent ceritinib in unresectable, refractory melanoma has been initiated based on this study although ceritinib may have greater potential in this disease when combined with MEK inhibitors.
Itraconazole
Itraconazole is an anti-fungal drug used in the treatment of systemic and superficial fungal infections that inhibits lanosterol-14α-demethylase, an enzyme involved in fungal membrane synthesis312; 313. While itraconazole’s inhibitory potential is much higher for fungal 14α-demethylase, it also inhibits human 14α-demethylase (involved in the biosynthesis of cholesterol) and this is believed to be involved in its reported anti-angiogenesis activity314. As an anticancer off-target effect, on the other hand, itraconazole has been shown through a phenotypic screen to block hedgehog pathway signaling by inhibiting smoothened (SMO)315. This activity seems to be different than that shown by other SMO inhibitors such as cyclopamine, suggesting a possibility for designing hedgehog inhibitor combination strategies although synergy with cisplatin has also been described316; 317. Multiple clinical studies of itraconazole are currently ongoing in various malignancies and there are several reviews that nicely summarize the current status of itraconazole repurposing in cancer312; 318; 319. Notably, an open-label, exploratory phase II trial of oral itraconazole has reported activity for the treatment of basal cell carcinoma320, but there have been also reports with significant, particularly cardiovascular, toxicities, which may limit itraconazole’s utility in malignant diseases320–322.
Disulfiram
Disulfiram was originally approved in the 1950’s to treat alcoholism. Inhibition of aldehyde dehydrogenase by disulfiram leads to accumulation of acetaldehyde, causing severe “hangover” symptoms upon alcohol ingestion323. Many theories have been suggested for the anticancer effects seen in patients treated with disulfiram, but in particular it has been proposed to be a proteasome inhibitor through a cell-based high-content screen323; 324. Along these lines, Skrott et al. showed that the active metabolite of disulfiram, the ditiocarb–copper complex, is binding and inhibiting NPL4, a p97 segregase adaptor, which functions upstream of the proteasome, thus leading to several overlapping, but also unique, properties compared to “true” proteasome inhibitors325. Disulfiram is currently being pursued for its anticancer properties in several active clinical trials and knowing its true target is expected to assist in developing mechanistic biomarkers for patient selection.
Chloroquine
In use since around 1940 to treat malaria, chloroquine was first tested as an anticancer agent for glioblastoma multiforme (GBM) in the late 1990’s. It has been in several clinical trials since then for various cancers as a single agent and in combination with chemotherapeutic drugs326; 327. Different from its use as an antimalarial, which is based on its ability to bind heme inside the parasitic food processing system and thereby blocking hemoglobin digestion, chloroquine has been used as an autophagy inhibitor in the context of cancer326; 327. Until recently, however, the actual molecular target of chloroquine and its related drugs, such as hydroxychloroquine and dimeric chloroquine analogues, was unknown. In 2017, using a chemical proteomics approach Rebecca et al. reported that palmitoyl-protein thioesterase 1 (PPT1), a protein involved in lysosomal degradation, may be that (off-)target328; 329. Importantly, they found that dimeric chloroquine analogues have better lysosomal inhibitory properties, blocking autophagy, but also inhibiting lysosomal-driven mTORC1 signaling328–330. It will be interesting to see how knowledge of the direct cancer target of chloroquines will be utilized in further development of these compounds for potentially biomarker-guided clinical trials.
Artemisinin
Artemisinin is a natural product isolated from the sweet wormwood plant, which has been used in Chinese traditional medicine to treat malaria for thousands of years. Artemisinin as the active compound was discovered in the early 1970’s by Tu Youyou, who was co-awarded the 2015 Nobel Prize in Medicine for this discovery331; 332. Most people believe that the antimalarial effects of artemisinins are due to the production of reactive oxygen species (ROS) derived from the degradation of the endoperoxide bridge, and this theory has also been put forward, at least partly, for artemisinin’s antitumor properties331; 333. Challenging this hypothesis, Gotsbacher et al. recently used a reverse chemical proteomics screen and identified a direct molecular target for artemisinin, namely the BCL-2 antagonist of cell death promoter BAD334. Since BAD is sequestered and inhibited upon phosphorylation, the ability of artemisinin to reduce phosphorylation of BAD, thereby increasing its pro-apoptotic activity, may allow for combination with other drugs that target the anti-apoptotic program prevalent in cancer cells334. Several clinical trials are currently active pursuing artemisinin (artesunate) activity in various cancers, such as colorectal cancer.
Sulfasalazine
Sulfasalazine is an anti-inflammatory drug used in inflammatory bowel disease and rheumatoid arthritis. In 2011, Chidley et al. discovered through a yeast three-hybrid target screen that the molecular target inhibited by sulfasalazine is sepiapterin reductase (SPR)335; 336. This protein is involved in the synthesis of the cofactor tetrahydrobiopterin (BH4), which is increased, for instance, in inflammation and pain337; 338. This pathway is also upregulated in neuroblastoma339. Although SPR is most likely also driving activity in the original indication and therefore strictly speaking can probably not be considered a sulfasalazine “off-target”, knowledge of the specific target may lead to more appropriate, and therefore more successful, development of sulfasalazine in cancer clinical trials, which are currently ongoing for instance in the pain management associated with breast cancer.
4. Conclusions
Having discussed various off-target-based repurposing examples in detail, it is worth extracting some specific aspects that can inform future repurposing efforts by highlighting opportunities and helping to avoid pitfalls. So, what can we learn from these cases? First, it should be noted that despite the stigma associated with off-targets and the initial cringe with which many scientists, specifically in the field of drug discovery, react, the end results from such efforts can be far from incremental, marginal or detrimental. Rather, they can have a profound positive impact, obviously on the lives of cancer patients who may not have otherwise effective therapies available, but also conceptually. Successful off-target-based repurposing can lead to elucidation of new mechanisms in cancer biology, inspire entirely new research directions and drug development programs or rekindle interest in existing fields by giving them a new spin25. This is best illustrated by crizotinib’s success in ALK- and ROS1-fusion positive NSCLC, which given the significant survival benefits achieved by crizotinib allowed the field to tackle acquired drug resistance in these tumors as a means to further prolong patient survival; a challenge that was in turn successfully met by the scientific and pharmaceutical community and in record time led to the development of several next-generation ALK and ROS1 inhibitors that have received regulatory approval, namely ceritinib, alectinib, brigatinib and lorlatinib for ALK-fusion positive, and most recently entrectinib for ROS1-fusion positive NSCLC. It is fairly safe to say that these accomplishments furthermore inspired new drug development efforts for other oncogenic fusion kinases, such as those involving RET and NTRK family kinases, thereby leading to the recent approval of larotrectinib and entrectinib for NTRK-fusion driven cancers. Likewise, the elucidation of thalidomide’s MoA as a consequence of major repurposing efforts and its employment as a targeted protein degrader have fueled the PROTAC field and led to nothing short of a surge in drug development programs that exploit this concept. It is also noteworthy that some drugs, for instance crizotinib and midostaurin, were repurposed and received approval due to their abilities to inhibit particular off-targets, not their cognate targets, thus highlighting the potential relevance of specifically elucidating off-target effects62; 87. Moreover, as demonstrated by dasatinib (BTK) and thalidomide (CRBN), elucidating the mechanisms underlying (off-target based) toxic drug side effects can lead to new repurposing initiatives25; 87.
These examples demonstrate that it is important to develop a detailed understanding of the targets and cellular MoAs of drugs62. While this is essentially always true, for off-target based repurposing approaches it is particularly critical to evaluate and functionally validate a drug-target interaction also in the correct biological context to ascertain that the off-target is inhibited potently enough and at the same time is a strong enough disease driver. This aspect may underlie the challenges encountered with erlotinib in JAK2-mutant AML or with imatinib in the context of breast cancer expressing KIT or PDGFRβ, the latter of which are validated imatinib targets with demonstrated clinical relevance when mutated for instance in GIST. This emphasizes that, as would be expected, the mechanism of oncogenic activation of a target is highly relevant and that gene fusions, mutations, gene amplification and (over)expression are not necessarily of equal importance across different cancers. In addition, some drugs display good potency for overexpressed or amplified wild-type targets, but not certain mutant forms of these, or vice versa. This is nicely exemplified by dasatinib and axitinib, which have opposite preferences for wild-type BCR-ABL and its T315I gatekeeper mutant, and by imatinib and midostaurin with regard to wild-type and D816V KIT.
Like with targeted therapies in general, it is furthermore apparent that drug repurposing is most successful when a predictive biomarker is available to assist in directing the drug to the right patient population62; 340. Of particularly high utility are genetic biomarkers, such as the presence of EML4-ALK translocations in NSCLC, activating KIT mutations in GIST or FLT3-ITD mutations in AML for indicating the use of crizotinib, imatinib and midostaurin, respectively. Although such markers may still not be sufficient to guarantee success in every case, as seen with erlotinib in JAK2-mutant AML and dasatinib in DDR2-mutant NSCLC for instance due to compensatory signaling, development of a biomarker in the context of drug repurposing is likely going to increase the chance for success of such effort. As the best predictive biomarkers have a strong mechanistic link, this again underscores the importance of obtaining detailed knowledge of the drug MoA and relevant target(s) and it will be interesting to follow future advances for instance with regard to repurposing of chloroquine or artemisinin now that specific drug targets of these have been identified in cancer cells.
The example of dasatinib repurposing for DDR2-mutant lung cancer, and similarly of tozasertib in BCR-ABL gatekeeper mutant CML, also shows that the potency of a drug for an off-target of interest has to be carefully considered in the context of clinically tolerable drug concentrations and drug toxicity, particularly as drugs have not been optimized for inhibition of off-targets and thus will often show weaker potency for these62. However, despite the ultimate failure of tozasertib, these efforts were not futile as the preclinical and clinical studies with tozasertib provided important proof of concept for efficacy and feasibility of targeting the BCR-ABL gatekeeper mutant and thus significantly supported the successful development of ponatinib, which currently constitutes the drug of last resort for CML patients. Cross-fertilization is also apparent for the development of ibrutinib and dasatinib as BTK inhibitors in hematological malignancies. Furthermore, if a drug is already approved for a different kind of cancer, like in the case of ibrutinib repurposing for lung or breast cancer, there will be already significant clinical expertise for handling its toxic side effects. However, as was seen with the curious case of desipramine in SCLC, there may be unique and unforeseeably low thresholds or potentially even different manifestations of toxicity in the context of a new disease25; 29.
The obvious solution for overcoming suboptimal potency against cancer-relevant off-targets is to replace the current drug with one that is known to have higher potency for this target. Unfortunately, such a compound is often not available. An alternative strategy that has been applied with some success to address this issue has been the design of drug combination therapies that provide a second hit either downstream of the relevant off-target or in a parallel pathway that contributes to oncogenic signaling or is involved in adaptive resistance through compensatory signaling. Notably, midostaurin has received regulatory approval for FLT3-ITD positive AML only in combination with conventional chemotherapy. Similarly, the combination of axitinib as a BCR-ABLT315I inhibitor with bosutinib is currently under clinical investigation for CML and a clinical trial of ceritinib with docetaxel, which enhances ceritinib’s polypharmacology-based efficacy, is ongoing in EML4-ALK-negative NSCLC.
The latter example also illustrates another, non-scientific, but equally critical factor for being able to successfully repurpose drugs, which is timing. As ceritinib had only relatively shortly before received regulatory approval for EML4-ALK-positive NSCLC, the intended indication, and thus secured a revenue flow, the pharmaceutical partner, in this case Novartis, was at that time interested in exploring and sponsoring additional therapeutic opportunities with ceritinib. This would have been unlikely to be of similar interest, if ceritinib had not received its initial approval yet or, conversely, if its patent protection period had been approaching its end. This latter aspect undoubtedly also facilitated the success of crizotinib as an ALK inhibitor considering that crizotinib drug development as a MET inhibitor was already in full swing when the EML4-ALK translocation was discovered as an oncogenic driver in NSCLC. It is remarkable, though, that Pfizer as the pharmaceutical company developing crizotinib, committed to exploring ALK-positive NSCLC already before crizotinib was approved, to some extent certainly also a testament to the strong validation of EML4-ALK as an oncogene. On the other hand, repurposing of midostaurin for EGFR-mutant NSCLC, which has developed resistance to EGFR TKI through acquisition of the T790M gatekeeper mutation, was not carried forward despite an at that time important unmet clinical need. This was probably due to the fact that midostaurin had not yet received its initial regulatory approval while at the same time its clinical development had taken unusually long thereby shortening its remaining patent life, factors that together would shift the risk versus potential benefit ratio and make an additional investment less favorable. In addition, there may have been a conflict with the pharmaceutical’s internal drug development pipeline, which contained nazartinib, a different (and newer) EGFR gatekeeper inhibitor. These cases demonstrate some of many, sometimes conflicting and often not transparent factors23; 25, which determine how much interest a pharmaceutical company can afford to pay to a new repurposing opportunity.
What these considerations furthermore highlight is that successful drug repurposing, be it off- or on-target, is a major team effort that involves multiple players with a spectrum of expertise23; 25. This is true already for the preclinical studies, which often start with a screen and continue with extensive phenotypic and mechanistic in vitro and in vivo validation before clinical translation can be considered. At this stage collaboration with pharmaceutical partners and their willingness/ability to sponsor clinical trials becomes critical as the costs for such trials usually exceed the budget of academic investigators, but also as they possess detailed knowledge of their drugs, for instance with regard to DMPK properties, side effects, administration and dosing schedules, which may not be available to the public. In addition, the repurposing approaches that are most successful involve clinician or physician-scientist collaborators or principal investigators, preferably at an institution with the staff and infrastructure to perform clinical studies. They have the necessary disease expertise, including detailed knowledge of the clinical needs and challenges, and access to the appropriate patient population and thus can evaluate the translational potential of any drug repurposing opportunity much better than a basic scientist and can point a project in a more impactful direction. Thus, the earlier a clinician is being involved in such an effort, the higher the potential for successful translation. Beyond this, clinicians often have established relationships with pharmaceutical companies through medical science liaisons whose job it is specifically to ease these interactions and promote clinical collaborations. Thus, their involvement can also provide access to communication channels, which can greatly facilitate collaboration between academic and pharmaceutical partners.
In summary, we here provide an overview of the rationale, methods applied and specific examples for off-target based repurposing in cancer. In doing so we furthermore discussed multiple scientific and non-scientific factors that either positively or negatively influence this process, some of which requiring careful consideration particularly for off-target based approaches. However, we hope we were also able to illustrate that off-targets should not just be viewed as potential causes of toxic side effects, but that off-target based drug repurposing efforts can be highly transformative and reveal novel or deepen our understanding of existing cancer biology mechanisms, alleviate the financial strain on pharmaceutical industry and, above all, provide urgently required new therapeutic approaches for cancer patients with previously unmet medical needs.
Acknowledgements
This work was supported by the National Institutes of Health (NIH)/National Cancer Institute (NCI) (awards R01 CA181746 and R01 CA219347) and the V Foundation.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of Interest
The authors declare no competing interests.
References
- 1.Chin L, Andersen JN, and Futreal PA (2011). Cancer genomics: from discovery science to personalized medicine. Nat Med 17, 297–303. [DOI] [PubMed] [Google Scholar]
- 2.Collins FS, and Varmus H (2015). A new initiative on precision medicine. N Engl J Med 372, 793–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ginsburg GS, and Willard HF (2009). Genomic and personalized medicine: foundations and applications. Transl Res 154, 277–287. [DOI] [PubMed] [Google Scholar]
- 4.Hamburg MA, and Collins FS (2010). The path to personalized medicine. N Engl J Med 363, 301–304. [DOI] [PubMed] [Google Scholar]
- 5.Baselga J, Norton L, Albanell J, Kim YM, and Mendelsohn J (1998). Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Res 58, 2825–2831. [PubMed] [Google Scholar]
- 6.Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, Druker BJ, and Lydon NB (1996). Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res 56, 100–104. [PubMed] [Google Scholar]
- 7.Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S, Zimmermann J, and Lydon NB (1996). Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med 2, 561–566. [DOI] [PubMed] [Google Scholar]
- 8.Pegram MD, Lipton A, Hayes DF, Weber BL, Baselga JM, Tripathy D, Baly D, Baughman SA, Twaddell T, Glaspy JA, et al. (1998). Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. J Clin Oncol 16, 2659–2671. [DOI] [PubMed] [Google Scholar]
- 9.Mardis ER (2008). The impact of next-generation sequencing technology on genetics. Trends Genet 24, 133–141. [DOI] [PubMed] [Google Scholar]
- 10.Metzker ML (2010). Sequencing technologies - the next generation. Nature reviews Genetics 11, 31–46. [DOI] [PubMed] [Google Scholar]
- 11.Ozsolak F, and Milos PM (2011). RNA sequencing: advances, challenges and opportunities. Nature reviews Genetics 12, 87–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Schuster SC (2008). Next-generation sequencing transforms today’s biology. Nat Methods 5, 16–18. [DOI] [PubMed] [Google Scholar]
- 13.Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, Srinivasan P, Gao J, Chakravarty D, Devlin SM, et al. (2017). Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 23, 703–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cancer Genome Atlas Research, N., Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C, and Stuart JM (2013). The Cancer Genome Atlas Pan-Cancer analysis project. Nature genetics 45, 1113–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, et al. (2007). Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Meyerson M, Gabriel S, and Getz G (2010). Advances in understanding cancer genomes through second-generation sequencing. Nature reviews Genetics 11, 685–696. [DOI] [PubMed] [Google Scholar]
- 17.Engelman JA, and Settleman J (2008). Acquired resistance to tyrosine kinase inhibitors during cancer therapy. Curr Opin Genet Dev 18, 73–79. [DOI] [PubMed] [Google Scholar]
- 18.Lovly CM, and Shaw AT (2014). Molecular pathways: resistance to kinase inhibitors and implications for therapeutic strategies. Clin Cancer Res 20, 2249–2256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Singh A, and Settleman J (2010). EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, and Sawyers CL (2001). Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 293, 876–880. [DOI] [PubMed] [Google Scholar]
- 21.Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, et al. (2010). Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 18, 683–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, Pochanard P, Kehoe SM, Johannessen CM, Macconaill LE, Hahn WC, et al. (2011). Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol 29, 3085–3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cha Y, Erez T, Reynolds IJ, Kumar D, Ross J, Koytiger G, Kusko R, Zeskind B, Risso S, Kagan E, et al. (2018). Drug repurposing from the perspective of pharmaceutical companies. Br J Pharmacol 175, 168–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Papapetropoulos A, and Szabo C (2018). Inventing new therapies without reinventing the wheel: the power of drug repurposing. Br J Pharmacol 175, 165–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, Doig A, Guilliams T, Latimer J, McNamee C, et al. (2019). Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov 18, 41–58. [DOI] [PubMed] [Google Scholar]
- 26.Khanna I (2012). Drug discovery in pharmaceutical industry: productivity challenges and trends. Drug Discov Today 17, 1088–1102. [DOI] [PubMed] [Google Scholar]
- 27.Ashburn TT, and Thor KB (2004). Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov 3, 673–683. [DOI] [PubMed] [Google Scholar]
- 28.Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, and Schutjens MH (2015). Drug repositioning and repurposing: terminology and definitions in literature. Drug Discov Today 20, 1027–1034. [DOI] [PubMed] [Google Scholar]
- 29.Wurth R, Thellung S, Bajetto A, Mazzanti M, Florio T, and Barbieri F (2016). Drug-repositioning opportunities for cancer therapy: novel molecular targets for known compounds. Drug Discov Today 21, 190–199. [DOI] [PubMed] [Google Scholar]
- 30.Shih HP, Zhang X, and Aronov AM (2018). Drug discovery effectiveness from the standpoint of therapeutic mechanisms and indications. Nat Rev Drug Discov 17, 19–33. [DOI] [PubMed] [Google Scholar]
- 31.Bertolini F, Sukhatme VP, and Bouche G (2015). Drug repurposing in oncology--patient and health systems opportunities. Nat Rev Clin Oncol 12, 732–742. [DOI] [PubMed] [Google Scholar]
- 32.Gns HS, Gr S, Murahari M, and Krishnamurthy M (2019). An update on Drug Repurposing: Rewritten saga of the drug’s fate. Biomed Pharmacother 110, 700–716. [DOI] [PubMed] [Google Scholar]
- 33.Ilmer M, Westphalen CB, Niess H, D’Haese JG, Angele MK, Werner J, and Renz BW (2019). Repurposed Drugs in Pancreatic Ductal Adenocarcinoma: An Update. Cancer J 25, 134–138. [DOI] [PubMed] [Google Scholar]
- 34.Murray JC, and Levy B (2019). Repurposed Drugs Trials by Cancer Type: Lung Cancer. Cancer J 25, 127–133. [DOI] [PubMed] [Google Scholar]
- 35.Pantziarka P, Verbaanderd C, Sukhatme V, Rica Capistrano I, Crispino S, Gyawali B, Rooman I, Van Nuffel AM, Meheus L, Sukhatme VP, et al. (2018). ReDO_DB: the repurposing drugs in oncology database. Ecancermedicalscience 12, 886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Allison M (2012). NCATS launches drug repurposing program. Nat Biotechnol 30, 571–572. [DOI] [PubMed] [Google Scholar]
- 37.Collins FS (2011). Reengineering translational science: the time is right. Sci Transl Med 3, 90cm17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cho D, Signoretti S, Regan M, Mier JW, and Atkins MB (2007). The role of mammalian target of rapamycin inhibitors in the treatment of advanced renal cancer. Clin Cancer Res 13, 758s–763s. [DOI] [PubMed] [Google Scholar]
- 39.Luan FL, Ding R, Sharma VK, Chon WJ, Lagman M, and Suthanthiran M (2003). Rapamycin is an effective inhibitor of human renal cancer metastasis. Kidney Int 63, 917–926. [DOI] [PubMed] [Google Scholar]
- 40.Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, and Morris AD (2005). Metformin and reduced risk of cancer in diabetic patients. BMJ 330, 1304–1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sandler RS, Halabi S, Baron JA, Budinger S, Paskett E, Keresztes R, Petrelli N, Pipas JM, Karp DD, Loprinzi CL, et al. (2003). A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. N Engl J Med 348, 883–890. [DOI] [PubMed] [Google Scholar]
- 42.Hogarty MD, Norris MD, Davis K, Liu X, Evageliou NF, Hayes CS, Pawel B, Guo R, Zhao H, Sekyere E, et al. (2008). ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer Res 68, 9735–9745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Rounbehler RJ, Li W, Hall MA, Yang C, Fallahi M, and Cleveland JL (2009). Targeting ornithine decarboxylase impairs development of MYCN-amplified neuroblastoma. Cancer Res 69, 547–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Overington JP, Al-Lazikani B, and Hopkins AL (2006). How many drug targets are there? Nat Rev Drug Discov 5, 993–996. [DOI] [PubMed] [Google Scholar]
- 45.Anastassiadis T, Deacon SW, Devarajan K, Ma H, and Peterson JR (2011). Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat Biotechnol 29, 1039–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, Klevernic I, Arthur JS, Alessi DR, and Cohen P (2007). The selectivity of protein kinase inhibitors: a further update. Biochem J 408, 297–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Davies SP, Reddy H, Caivano M, and Cohen P (2000). Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351, 95–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Davis MI, Hunt JP, Herrgard S, Ciceri P, Wodicka LM, Pallares G, Hocker M, Treiber DK, and Zarrinkar PP (2011). Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol 29, 1046–1051. [DOI] [PubMed] [Google Scholar]
- 49.Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT, et al. (2008). A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol 26, 127–132. [DOI] [PubMed] [Google Scholar]
- 50.Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB, et al. (2003). In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 9, 327–337. [PubMed] [Google Scholar]
- 51.Yakes FM, Chen J, Tan J, Yamaguchi K, Shi Y, Yu P, Qian F, Chu F, Bentzien F, Cancilla B, et al. (2011). Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther 10, 2298–2308. [DOI] [PubMed] [Google Scholar]
- 52.Zhang Y, Guessous F, Kofman A, Schiff D, and Abounader R (2010). XL-184, a MET, VEGFR-2 and RET kinase inhibitor for the treatment of thyroid cancer, glioblastoma multiforme and NSCLC. IDrugs 13, 112–121. [PMC free article] [PubMed] [Google Scholar]
- 53.Blaugrund JE, Johns MM Jr., Eby YJ, Ball DW, Baylin SB, Hruban RH, and Sidransky D (1994). RET proto-oncogene mutations in inherited and sporadic medullary thyroid cancer. Human molecular genetics 3, 1895–1897. [DOI] [PubMed] [Google Scholar]
- 54.Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, Kawano K, Hanada M, Kurata A, Takeda M, et al. (1998). Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279, 577–580. [DOI] [PubMed] [Google Scholar]
- 55.Atkins M, Jones CA, and Kirkpatrick P (2006). Sunitinib maleate. Nat Rev Drug Discov 5, 279–280. [DOI] [PubMed] [Google Scholar]
- 56.Chow LQ, and Eckhardt SG (2007). Sunitinib: from rational design to clinical efficacy. J Clin Oncol 25, 884–896. [DOI] [PubMed] [Google Scholar]
- 57.Elisei R, Schlumberger MJ, Muller SP, Schoffski P, Brose MS, Shah MH, Licitra L, Jarzab B, Medvedev V, Kreissl MC, et al. (2013). Cabozantinib in progressive medullary thyroid cancer. J Clin Oncol 31, 3639–3646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kurzrock R, Sherman SI, Ball DW, Forastiere AA, Cohen RB, Mehra R, Pfister DG, Cohen EE, Janisch L, Nauling F, et al. (2011). Activity of XL184 (Cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J Clin Oncol 29, 2660–2666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Johnson TW, Richardson PF, Bailey S, Brooun A, Burke BJ, Collins MR, Cui JJ, Deal JG, Deng YL, Dinh D, et al. (2014). Discovery of (10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(m etheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a macrocyclic inhibitor of anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) with preclinical brain exposure and broad-spectrum potency against ALK-resistant mutations. J Med Chem 57, 4720–4744. [DOI] [PubMed] [Google Scholar]
- 60.Zou HY, Li Q, Engstrom LD, West M, Appleman V, Wong KA, McTigue M, Deng YL, Liu W, Brooun A, et al. (2015). PF-06463922 is a potent and selective next-generation ROS1/ALK inhibitor capable of blocking crizotinib-resistant ROS1 mutations. Proc Natl Acad Sci U S A 112, 3493–3498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hopkins AL (2008). Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol 4, 682–690. [DOI] [PubMed] [Google Scholar]
- 62.Antolin AA, Workman P, Mestres J, and Al-Lazikani B (2016). Polypharmacology in Precision Oncology: Current Applications and Future Prospects. Current pharmaceutical design 22, 6935–6945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Boran AD, and Iyengar R (2010). Systems approaches to polypharmacology and drug discovery. Curr Opin Drug Discov Devel 13, 297–309. [PMC free article] [PubMed] [Google Scholar]
- 64.Csermely P, Agoston V, and Pongor S (2005). The efficiency of multi-target drugs: the network approach might help drug design. Trends Pharmacol Sci 26, 178–182. [DOI] [PubMed] [Google Scholar]
- 65.Greaves M (2015). Evolutionary determinants of cancer. Cancer Discov 5, 806–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Liu YY, Slotine JJ, and Barabasi AL (2011). Controllability of complex networks. Nature 473, 167–173. [DOI] [PubMed] [Google Scholar]
- 67.Workman P, Al-Lazikani B, and Clarke PA (2013). Genome-based cancer therapeutics: targets, kinase drug resistance and future strategies for precision oncology. Curr Opin Pharmacol 13, 486–496. [DOI] [PubMed] [Google Scholar]
- 68.Davies KD, Mahale S, Astling DP, Aisner DL, Le AT, Hinz TK, Vaishnavi A, Bunn PA Jr., Heasley LE, Tan AC, et al. (2013). Resistance to ROS1 inhibition mediated by EGFR pathway activation in non-small cell lung cancer. PloS one 8, e82236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sasaki T, Koivunen J, Ogino A, Yanagita M, Nikiforow S, Zheng W, Lathan C, Marcoux JP, Du J, Okuda K, et al. (2011). A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res 71, 6051–6060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Vaishnavi A, Schubert L, Rix U, Marek LA, Le AT, Keysar SB, Glogowska MJ, Smith MA, Kako S, Sumi NJ, et al. (2017). EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases. Cancer Res 77, 3551–3563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al. (2007). MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316, 1039–1043. [DOI] [PubMed] [Google Scholar]
- 72.Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, and Bernards R (2012). Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103. [DOI] [PubMed] [Google Scholar]
- 73.Janku F, Yap TA, and Meric-Bernstam F (2018). Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol 15, 273–291. [DOI] [PubMed] [Google Scholar]
- 74.Rozengurt E, Soares HP, and Sinnet-Smith J (2014). Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol Cancer Ther 13, 2477–2488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sarbassov DD, Guertin DA, Ali SM, and Sabatini DM (2005). Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307, 1098–1101. [DOI] [PubMed] [Google Scholar]
- 76.Barabasi AL, Gulbahce N, and Loscalzo J (2011). Network medicine: a network-based approach to human disease. Nature reviews Genetics 12, 56–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pujol A, Mosca R, Farres J, and Aloy P (2010). Unveiling the role of network and systems biology in drug discovery. Trends Pharmacol Sci 31, 115–123. [DOI] [PubMed] [Google Scholar]
- 78.Shah KN, Bhatt R, Rotow J, Rohrberg J, Olivas V, Wang VE, Hemmati G, Martins MM, Maynard A, Kuhn J, et al. (2019). Aurora kinase A drives the evolution of resistance to thirdgeneration EGFR inhibitors in lung cancer. Nat Med 25, 111–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, McDermott U, Azizian N, Zou L, Fischbach MA, et al. (2010). A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hata AN, Niederst MJ, Archibald HL, Gomez-Caraballo M, Siddiqui FM, Mulvey HE, Maruvka YE, Ji F, Bhang HE, Krishnamurthy Radhakrishna V, et al. (2016). Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat Med 22, 262–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ramirez M, Rajaram S, Steininger RJ, Osipchuk D, Roth MA, Morinishi LS, Evans L, Ji W, Hsu CH, Thurley K, et al. (2016). Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells. Nat Commun 7, 10690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Coxib, traditional, N.T.C., Bhala N, Emberson J, Merhi A, Abramson S, Arber N, Baron JA, Bombardier C, Cannon C, et al. (2013). Vascular and upper gastrointestinal effects of nonsteroidal anti-inflammatory drugs: meta-analyses of individual participant data from randomised trials. Lancet 382, 769–779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Maxwell SR, and Webb DJ (2005). COX-2 selective inhibitors--important lessons learned. Lancet 365, 449–451. [DOI] [PubMed] [Google Scholar]
- 84.Holohan C, Van Schaeybroeck S, Longley DB, and Johnston PG (2013). Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 13, 714–726. [DOI] [PubMed] [Google Scholar]
- 85.Palmer AC, and Sorger PK (2017). Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy. Cell 171, 1678–1691 e1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Sun W, Sanderson PE, and Zheng W (2016). Drug combination therapy increases successful drug repositioning. Drug Discov Today 21, 1189–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Anighoro A, Bajorath J, and Rastelli G (2014). Polypharmacology: challenges and opportunities in drug discovery. J Med Chem 57, 7874–7887. [DOI] [PubMed] [Google Scholar]
- 88.Knight ZA, Lin H, and Shokat KM (2010). Targeting the cancer kinome through polypharmacology. Nat Rev Cancer 10, 130–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Achenbach J, Tiikkainen P, Franke L, and Proschak E (2011). Computational tools for polypharmacology and repurposing. Future Med Chem 3, 961–968. [DOI] [PubMed] [Google Scholar]
- 90.Campillos M, Kuhn M, Gavin AC, Jensen LJ, and Bork P (2008). Drug target identification using side-effect similarity. Science 321, 263–266. [DOI] [PubMed] [Google Scholar]
- 91.Hieronymus H, Lamb J, Ross KN, Peng XP, Clement C, Rodina A, Nieto M, Du J, Stegmaier K, Raj SM, et al. (2006). Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell 10, 321–330. [DOI] [PubMed] [Google Scholar]
- 92.Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, et al. (2006). The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935. [DOI] [PubMed] [Google Scholar]
- 93.Subramanian A, Narayan R, Corsello SM, Peck DD, Natoli TE, Lu X, Gould J, Davis JF, Tubelli AA, Asiedu JK, et al. (2017). A Next Generation Connectivity Map: L1000 Platform and the First 1,000,000 Profiles. Cell 171, 1437–1452 e1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Fernandez-Torras A, Duran-Frigola M, and Aloy P (2019). Encircling the regions of the pharmacogenomic landscape that determine drug response. Genome Med 11, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gregori-Puigjane E, and Mestres J (2008). A ligand-based approach to mining the chemogenomic space of drugs. Comb Chem High Throughput Screen 11, 669–676. [DOI] [PubMed] [Google Scholar]
- 96.Paolini GV, Shapland RH, van Hoorn WP, Mason JS, and Hopkins AL (2006). Global mapping of pharmacological space. Nat Biotechnol 24, 805–815. [DOI] [PubMed] [Google Scholar]
- 97.Woo JH, Shimoni Y, Yang WS, Subramaniam P, Iyer A, Nicoletti P, Rodriguez Martinez M, Lopez G, Mattioli M, Realubit R, et al. (2015). Elucidating Compound Mechanism of Action by Network Perturbation Analysis. Cell 162, 441–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Keiser MJ, Roth BL, Armbruster BN, Ernsberger P, Irwin JJ, and Shoichet BK (2007). Relating protein pharmacology by ligand chemistry. Nat Biotechnol 25, 197–206. [DOI] [PubMed] [Google Scholar]
- 99.Keiser MJ, Setola V, Irwin JJ, Laggner C, Abbas AI, Hufeisen SJ, Jensen NH, Kuijer MB, Matos RC, Tran TB, et al. (2009). Predicting new molecular targets for known drugs. Nature 462, 175–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP, and Vikas P (2014). The Repurposing Drugs in Oncology (ReDO) Project. Ecancermedicalscience 8, 442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chen H, Engkvist O, Wang Y, Olivecrona M, and Blaschke T (2018). The rise of deep learning in drug discovery. Drug Discov Today 23, 1241–1250. [DOI] [PubMed] [Google Scholar]
- 102.Ekins S (2016). The Next Era: Deep Learning in Pharmaceutical Research. Pharm Res 33, 2594–2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Moffat JG, Rudolph J, and Bailey D (2014). Phenotypic screening in cancer drug discovery - past, present and future. Nat Rev Drug Discov 13, 588–602. [DOI] [PubMed] [Google Scholar]
- 104.Zheng W, Thorne N, and McKew JC (2013). Phenotypic screens as a renewed approach for drug discovery. Drug Discov Today 18, 1067–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Eggert US (2013). The why and how of phenotypic small-molecule screens. Nat Chem Biol 9, 206–209. [DOI] [PubMed] [Google Scholar]
- 106.Muellner MK, Mair B, Ibrahim Y, Kerzendorfer C, Lechtermann H, Trefzer C, Klepsch F, Muller AC, Leitner E, Macho-Maschler S, et al. (2015). Targeting a cell state common to triple-negative breast cancers. Mol Syst Biol 11, 789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Castoreno AB, Smurnyy Y, Torres AD, Vokes MS, Jones TR, Carpenter AE, and Eggert US (2010). Small molecules discovered in a pathway screen target the Rho pathway in cytokinesis. Nat Chem Biol 6, 457–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Gazdar AF, Girard L, Lockwood WW, Lam WL, and Minna JD (2010). Lung cancer cell lines as tools for biomedical discovery and research. J Natl Cancer Inst 102, 1310–1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Shoemaker RH, Monks A, Alley MC, Scudiero DA, Fine DL, McLemore TL, Abbott BJ, Paull KD, Mayo JG, and Boyd MR (1988). Development of human tumor cell line panels for use in disease-oriented drug screening. Prog Clin Biol Res 276, 265–286. [PubMed] [Google Scholar]
- 110.Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehar J, Kryukov GV, Sonkin D, et al. (2012). The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Basu A, Bodycombe NE, Cheah JH, Price EV, Liu K, Schaefer GI, Ebright RY, Stewart ML, Ito D, Wang S, et al. (2013). An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell 154, 1151–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, Greninger P, Thompson IR, Luo X, Soares J, et al. (2012). Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483, 570–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ghandi M, Huang FW, Jane-Valbuena J, Kryukov GV, Lo CC, McDonald ER 3rd, Barretina J, Gelfand ET, Bielski CM, Li H, et al. (2019). Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Holbeck SL, Collins JM, and Doroshow JH (2010). Analysis of Food and Drug Administration-approved anticancer agents in the NCI60 panel of human tumor cell lines. Mol Cancer Ther 9, 1451–1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.McDermott U, Sharma SV, Dowell L, Greninger P, Montagut C, Lamb J, Archibald H, Raudales R, Tam A, Lee D, et al. (2007). Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A 104, 19936–19941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Seashore-Ludlow B, Rees MG, Cheah JH, Cokol M, Price EV, Coletti ME, Jones V, Bodycombe NE, Soule CK, Gould J, et al. (2015). Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset. Cancer Discov 5, 1210–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Weinstein JN, Myers TG, O’Connor PM, Friend SH, Fornace AJ Jr., Kohn KW, Fojo T, Bates SE, Rubinstein LV, Anderson NL, et al. (1997). An information-intensive approach to the molecular pharmacology of cancer. Science 275, 343–349. [DOI] [PubMed] [Google Scholar]
- 118.Iorio F, Knijnenburg TA, Vis DJ, Bignell GR, Menden MP, Schubert M, Aben N, Goncalves E, Barthorpe S, Lightfoot H, et al. (2016). A Landscape of Pharmacogenomic Interactions in Cancer. Cell 166, 740–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Monks A, Zhao Y, Hose C, Hamed H, Krushkal J, Fang J, Sonkin D, Palmisano A, Polley EC, Fogli LK, et al. (2018). The NCI Transcriptional Pharmacodynamics Workbench: A Tool to Examine Dynamic Expression Profiling of Therapeutic Response in the NCI-60 Cell Line Panel. Cancer Res 78, 6807–6817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kota S, Hou S, Guerrant W, Madoux F, Troutman S, Fernandez-Vega V, Alekseeva N, Madala N, Scampavia L, Kissil J, et al. (2018). A novel three-dimensional high-throughput screening approach identifies inducers of a mutant KRAS selective lethal phenotype. Oncogene 37, 4372–4384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Liu X, Ory V, Chapman S, Yuan H, Albanese C, Kallakury B, Timofeeva OA, Nealon C, Dakic A, Simic V, et al. (2012). ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol 180, 599–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Quereda V, Hou S, Madoux F, Scampavia L, Spicer TP, and Duckett D (2018). A Cytotoxic Three-Dimensional-Spheroid, High-Throughput Assay Using Patient-Derived Glioma Stem Cells. SLAS Discov 23, 842–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kodack DP, Farago AF, Dastur A, Held MA, Dardaei L, Friboulet L, von Flotow F, Damon LJ, Lee D, Parks M, et al. (2017). Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care. Cell reports 21, 3298–3309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, Zhang C, Schnell C, Yang G, Zhang Y, et al. (2015). High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med 21, 1318–1325. [DOI] [PubMed] [Google Scholar]
- 125.Dar AC, Das TK, Shokat KM, and Cagan RL (2012). Chemical genetic discovery of targets and anti-targets for cancer polypharmacology. Nature 486, 80–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Kim WH, Shen H, Jung DW, and Williams DR (2016). Some leopards can change their spots: potential repositioning of stem cell reprogramming compounds as anti-cancer agents. Cell Biol Toxicol 32, 157–168. [DOI] [PubMed] [Google Scholar]
- 127.MacRae CA, and Peterson RT (2015). Zebrafish as tools for drug discovery. Nat Rev Drug Discov 14, 721–731. [DOI] [PubMed] [Google Scholar]
- 128.Murphey RD, Stern HM, Straub CT, and Zon LI (2006). A chemical genetic screen for cell cycle inhibitors in zebrafish embryos. Chemical biology & drug design 68, 213–219. [DOI] [PubMed] [Google Scholar]
- 129.White R, Rose K, and Zon L (2013). Zebrafish cancer: the state of the art and the path forward. Nat Rev Cancer 13, 624–636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Ctortecka C, Palve V, Kuenzi BM, Fang B, Sumi NJ, Izumi V, Novakova S, Kinose F, Remsing Rix LL, Haura EB, et al. (2018). Functional Proteomics and Deep Network Interrogation Reveal a Complex Mechanism of Action of Midostaurin in Lung Cancer Cells. Mol Cell Proteomics 17, 2434–2447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Kuenzi BM, Remsing Rix LL, Stewart PA, Fang B, Kinose F, Bryant AT, Boyle TA, Koomen JM, Haura EB, and Rix U (2017). Polypharmacology-based ceritinib repurposing using integrated functional proteomics. Nat Chem Biol 13, 1222–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Remsing Rix LL, Kuenzi BM, Luo Y, Remily-Wood E, Kinose F, Wright G, Li J, Koomen JM, Haura EB, Lawrence HR, et al. (2014). GSK3 Alpha and Beta Are New Functionally Relevant Targets of Tivantinib in Lung Cancer Cells. ACS chemical biology 9, 353–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Sumi NJ, Ctortecka C, Hu Q, Bryant AT, Fang B, Remsing Rix LL, Ayaz M, Kinose F, Welsh EA, Eschrich SA, et al. (2019). Divergent Polypharmacology-Driven Cellular Activity of Structurally Similar Multi-Kinase Inhibitors through Cumulative Effects on Individual Targets. Cell chemical biology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Nijman SM (2015). Functional genomics to uncover drug mechanism of action. Nat Chem Biol 11, 942–948. [DOI] [PubMed] [Google Scholar]
- 135.Schenone M, Dancik V, Wagner BK, and Clemons PA (2013). Target identification and mechanism of action in chemical biology and drug discovery. Nat Chem Biol 9, 232–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ziegler S, Pries V, Hedberg C, and Waldmann H (2013). Target identification for small bioactive molecules: finding the needle in the haystack. Angewandte Chemie 52, 2744–2792. [DOI] [PubMed] [Google Scholar]
- 137.MacBeath G, and Schreiber SL (2000). Printing proteins as microarrays for high-throughput function determination. Science 289, 1760–1763. [DOI] [PubMed] [Google Scholar]
- 138.Huang J, Zhu H, Haggarty SJ, Spring DR, Hwang H, Jin F, Snyder M, and Schreiber SL (2004). Finding new components of the target of rapamycin (TOR) signaling network through chemical genetics and proteome chips. Proc Natl Acad Sci U S A 101, 16594–16599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Rix U, and Superti-Furga G (2009). Target profiling of small molecules by chemical proteomics. Nat Chem Biol 5, 616–624. [DOI] [PubMed] [Google Scholar]
- 140.Barglow KT, and Cravatt BF (2007). Activity-based protein profiling for the functional annotation of enzymes. Nat Methods 4, 822–827. [DOI] [PubMed] [Google Scholar]
- 141.Weerapana E, Wang C, Simon GM, Richter F, Khare S, Dillon MB, Bachovchin DA, Mowen K, Baker D, and Cravatt BF (2010). Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Patricelli MP, Nomanbhoy TK, Wu J, Brown H, Zhou D, Zhang J, Jagannathan S, Aban A, Okerberg E, Herring C, et al. (2011). In situ kinase profiling reveals functionally relevant properties of native kinases. Chem Biol 18, 699–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tully SE, and Cravatt BF (2010). Activity-based probes that target functional subclasses of phospholipases in proteomes. J Am Chem Soc 132, 3264–3265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Bantscheff M, Eberhard D, Abraham Y, Bastuck S, Boesche M, Hobson S, Mathieson T, Perrin J, Raida M, Rau C, et al. (2007). Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat Biotechnol 25, 1035–1044. [DOI] [PubMed] [Google Scholar]
- 145.Klaeger S, Heinzlmeir S, Wilhelm M, Polzer H, Vick B, Koenig PA, Reinecke M, Ruprecht B, Petzoldt S, Meng C, et al. (2017). The target landscape of clinical kinase drugs. Science 358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Savitski MM, Reinhard FB, Franken H, Werner T, Savitski MF, Eberhard D, Martinez Molina D, Jafari R, Dovega RB, Klaeger S, et al. (2014). Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 346, 1255784. [DOI] [PubMed] [Google Scholar]
- 147.Sharma K, Weber C, Bairlein M, Greff Z, Keri G, Cox J, Olsen JV, and Daub H (2009). Proteomics strategy for quantitative protein interaction profiling in cell extracts. Nat Methods 6, 741–744. [DOI] [PubMed] [Google Scholar]
- 148.Godl K, Wissing J, Kurtenbach A, Habenberger P, Blencke S, Gutbrod H, Salassidis K, Stein-Gerlach M, Missio A, Cotten M, et al. (2003). An efficient proteomics method to identify the cellular targets of protein kinase inhibitors. Proc Natl Acad Sci U S A 100, 15434–15439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Huang SM, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, Charlat O, Wiellette E, Zhang Y, Wiessner S, et al. (2009). Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 461, 614–620. [DOI] [PubMed] [Google Scholar]
- 150.Knezevic CE, Wright G, Remsing Rix LL, Kim W, Kuenzi BM, Luo Y, Watters JM, Koomen JM, Haura EB, Monteiro AN, et al. (2016). Proteome-wide Profiling of Clinical PARP Inhibitors Reveals Compound-Specific Secondary Targets. Cell chemical biology 23, 1490–1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ong SE, Schenone M, Margolin AA, Li X, Do K, Doud MK, Mani DR, Kuai L, Wang X, Wood JL, et al. (2009). Identifying the proteins to which small-molecule probes and drugs bind in cells. Proc Natl Acad Sci U S A 106, 4617–4622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Cui JJ, Tran-Dube M, Shen H, Nambu M, Kung PP, Pairish M, Jia L, Meng J, Funk L, Botrous I, et al. (2011). Structure based drug design of crizotinib (PF-02341066), a potent and selective dual inhibitor of mesenchymal-epithelial transition factor (c-MET) kinase and anaplastic lymphoma kinase (ALK). J Med Chem 54, 6342–6363. [DOI] [PubMed] [Google Scholar]
- 153.Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung PP, et al. (2007). An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res 67, 4408–4417. [DOI] [PubMed] [Google Scholar]
- 154.Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, et al. (2007). Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 448, 561–566. [DOI] [PubMed] [Google Scholar]
- 155.McDermott U, Iafrate AJ, Gray NS, Shioda T, Classon M, Maheswaran S, Zhou W, Choi HG, Smith SL, Dowell L, et al. (2008). Genomic alterations of anaplastic lymphoma kinase may sensitize tumors to anaplastic lymphoma kinase inhibitors. Cancer Res 68, 3389–3395. [DOI] [PubMed] [Google Scholar]
- 156.Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, Riely GJ, Solomon B, Ou SH, Kim DW, et al. (2012). Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol 13, 1011–1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Janne PA, Costa DB, et al. (2010). Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 363, 1693–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Shaw AT, Kim DW, Nakagawa K, Seto T, Crino L, Ahn MJ, De Pas T, Besse B, Solomon BJ, Blackhall F, et al. (2013). Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 368, 2385–2394. [DOI] [PubMed] [Google Scholar]
- 159.Bergethon K, Shaw AT, Ou SH, Katayama R, Lovly CM, McDonald NT, Massion PP, Siwak-Tapp C, Gonzalez A, Fang R, et al. (2012). ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol 30, 863–870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, Nardone J, Lee K, Reeves C, Li Y, et al. (2007). Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131, 1190–1203. [DOI] [PubMed] [Google Scholar]
- 161.Davies KD, and Doebele RC (2013). Molecular pathways: ROS1 fusion proteins in cancer. Clin Cancer Res 19, 4040–4045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Schram AM, Chang MT, Jonsson P, and Drilon A (2017). Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol 14, 735–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Shaw AT, Ou SH, Bang YJ, Camidge DR, Solomon BJ, Salgia R, Riely GJ, Varella-Garcia M, Shapiro GI, Costa DB, et al. (2014). Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 371, 1963–1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Katayama R, Khan TM, Benes C, Lifshits E, Ebi H, Rivera VM, Shakespeare WC, Iafrate AJ, Engelman JA, and Shaw AT (2011). Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK. Proc Natl Acad Sci U S A 108, 7535–7540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Lin JJ, Riely GJ, and Shaw AT (2017). Targeting ALK: Precision Medicine Takes on Drug Resistance. Cancer Discov 7, 137–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Kantarjian H, Sawyers C, Hochhaus A, Guilhot F, Schiffer C, Gambacorti-Passerini C, Niederwieser D, Resta D, Capdeville R, Zoellner U, et al. (2002). Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med 346, 645–652. [DOI] [PubMed] [Google Scholar]
- 167.Mauro MJ, O’Dwyer M, Heinrich MC, and Druker BJ (2002). STI571: a paradigm of new agents for cancer therapeutics. J Clin Oncol 20, 325–334. [DOI] [PubMed] [Google Scholar]
- 168.Capdeville R, Buchdunger E, Zimmermann J, and Matter A (2002). Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 1, 493–502. [DOI] [PubMed] [Google Scholar]
- 169.Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, and Zigler AJ (2000). Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood 96, 925–932. [PubMed] [Google Scholar]
- 170.Demetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts PJ, Heinrich MC, Tuveson DA, Singer S, Janicek M, et al. (2002). Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 347, 472–480. [DOI] [PubMed] [Google Scholar]
- 171.Corless CL, Schroeder A, Griffith D, Town A, McGreevey L, Harrell P, Shiraga S, Bainbridge T, Morich J, and Heinrich MC (2005). PDGFRA mutations in gastrointestinal stromal tumors: frequency, spectrum and in vitro sensitivity to imatinib. J Clin Oncol 23, 5357–5364. [DOI] [PubMed] [Google Scholar]
- 172.Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, Joensuu H, McGreevey LS, Chen CJ, Van den Abbeele AD, Druker BJ, et al. (2003). Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 21, 4342–4349. [DOI] [PubMed] [Google Scholar]
- 173.Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, Pasquini R, Clark RE, Hochhaus A, Hughes TP, et al. (2010). Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med 362, 2251–2259. [DOI] [PubMed] [Google Scholar]
- 174.Blay JY, Shen L, Kang YK, Rutkowski P, Qin S, Nosov D, Wan D, Trent J, Srimuninnimit V, Papai Z, et al. (2015). Nilotinib versus imatinib as first-line therapy for patients with unresectable or metastatic gastrointestinal stromal tumours (ENESTg1): a randomised phase 3 trial. Lancet Oncol 16, 550–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Yam C, Murthy RK, Rauch GM, Murray JL, Walters RS, Valero V, Brewster AM, Bast RC Jr., Booser DJ, Giordano SH, et al. (2018). A phase II study of imatinib mesylate and letrozole in patients with hormone receptor-positive metastatic breast cancer expressing c-kit or PDGFR-beta. Investigational new drugs 36, 1103–1109. [DOI] [PubMed] [Google Scholar]
- 176.Guo J, Si L, Kong Y, Flaherty KT, Xu X, Zhu Y, Corless CL, Li L, Li H, Sheng X, et al. (2011). Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol 29, 2904–2909. [DOI] [PubMed] [Google Scholar]
- 177.Wei X, Mao L, Chi Z, Sheng X, Cui C, Kong Y, Dai J, Wang X, Li S, Tang B, et al. (2019). Efficacy Evaluation of Imatinib for the Treatment of Melanoma: Evidence From a Retrospective Study. Oncol Res 27, 495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Hodi FS, Corless CL, Giobbie-Hurder A, Fletcher JA, Zhu M, Marino-Enriquez A, Friedlander P, Gonzalez R, Weber JS, Gajewski TF, et al. (2013). Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol 31, 3182–3190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Fabbro D, Buchdunger E, Wood J, Mestan J, Hofmann F, Ferrari S, Mett H, O’Reilly T, and Meyer T (1999). Inhibitors of protein kinases: CGP 41251, a protein kinase inhibitor with potential as an anticancer agent. Pharmacology & therapeutics 82, 293–301. [DOI] [PubMed] [Google Scholar]
- 180.Fabbro D, Ruetz S, Bodis S, Pruschy M, Csermak K, Man A, Campochiaro P, Wood J, O’Reilly T, and Meyer T (2000). PKC412--a protein kinase inhibitor with a broad therapeutic potential. Anticancer Drug Des 15, 17–28. [PubMed] [Google Scholar]
- 181.Stone RM, Manley PW, Larson RA, and Capdeville R (2018). Midostaurin: its odyssey from discovery to approval for treating acute myeloid leukemia and advanced systemic mastocytosis. Blood Adv 2, 444–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Weisberg E, Boulton C, Kelly LM, Manley P, Fabbro D, Meyer T, Gilliland DG, and Griffin JD (2002). Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell 1, 433–443. [DOI] [PubMed] [Google Scholar]
- 183.Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, Sonoda Y, Fujimoto T, and Misawa S (1996). Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 10, 1911–1918. [PubMed] [Google Scholar]
- 184.Stone RM, DeAngelo DJ, Klimek V, Galinsky I, Estey E, Nimer SD, Grandin W, Lebwohl D, Wang Y, Cohen P, et al. (2005). Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood 105, 54–60. [DOI] [PubMed] [Google Scholar]
- 185.Levis M (2017). Midostaurin approved for FLT3-mutated AML. Blood 129, 3403–3406. [DOI] [PubMed] [Google Scholar]
- 186.Stone RM, Fischer T, Paquette R, Schiller G, Schiffer CA, Ehninger G, Cortes J, Kantarjian HM, DeAngelo DJ, Huntsman-Labed A, et al. (2012). Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia. Leukemia 26, 2061–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Cullinane C, Natoli A, Hui Y, Conus N, Jackson S, Bruggen J, Manley PW, and McArthur GA (2010). Preclinical evaluation of nilotinib efficacy in an imatinib-resistant KIT-driven tumor model. Mol Cancer Ther 9, 1461–1468. [DOI] [PubMed] [Google Scholar]
- 188.Gotlib J, Berube C, Growney JD, Chen CC, George TI, Williams C, Kajiguchi T, Ruan J, Lilleberg SL, Durocher JA, et al. (2005). Activity of the tyrosine kinase inhibitor PKC412 in a patient with mast cell leukemia with the D816V KIT mutation. Blood 106, 2865–2870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.DeAngelo DJ, George TI, Linder A, Langford C, Perkins C, Ma J, Westervelt P, Merker JD, Berube C, Coutre S, et al. (2018). Efficacy and safety of midostaurin in patients with advanced systemic mastocytosis: 10-year median follow-up of a phase II trial. Leukemia 32, 470–478. [DOI] [PubMed] [Google Scholar]
- 190.Gotlib J, Kluin-Nelemans HC, George TI, Akin C, Sotlar K, Hermine O, Awan FT, Hexner E, Mauro MJ, Sternberg DW, et al. (2016). Efficacy and Safety of Midostaurin in Advanced Systemic Mastocytosis. N Engl J Med 374, 2530–2541. [DOI] [PubMed] [Google Scholar]
- 191.Kasamon YL, Ko CW, Subramaniam S, Ma L, Yang Y, Nie L, Shord S, Przepiorka D, Farrell AT, McKee AE, et al. (2018). FDA Approval Summary: Midostaurin for the Treatment of Advanced Systemic Mastocytosis. Oncologist 23, 1511–1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Lee HJ, Schaefer G, Heffron TP, Shao L, Ye X, Sideris S, Malek S, Chan E, Merchant M, La H, et al. (2013). Noncovalent wild-type-sparing inhibitors of EGFR T790M. Cancer Discov 3, 168–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Li J, Bennett K, Stukalov A, Fang B, Zhang G, Yoshida T, Okamoto I, Kim JY, Song L, Bai Y, et al. (2013). Perturbation of the mutated EGFR interactome identifies vulnerabilities and resistance mechanisms. Mol Syst Biol 9, 705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Eleutherakis-Papaiakovou V, Bamias A, and Dimopoulos MA (2004). Thalidomide in cancer medicine. Ann Oncol 15, 1151–1160. [DOI] [PubMed] [Google Scholar]
- 195.Franks ME, Macpherson GR, and Figg WD (2004). Thalidomide. Lancet 363, 1802–1811. [DOI] [PubMed] [Google Scholar]
- 196.Lenz W, and Knapp K (1962). [Thalidomide embryopathy]. Dtsch Med Wochenschr 87, 1232–1242. [DOI] [PubMed] [Google Scholar]
- 197.Mellin GW, and Katzenstein M (1962). The saga of thalidomide. Neuropathy to embryopathy, with case reports of congenital anomalies. N Engl J Med 267, 1184–1192 contd. [DOI] [PubMed] [Google Scholar]
- 198.Lenz W (1988). A short history of thalidomide embryopathy. Teratology 38, 203–215. [DOI] [PubMed] [Google Scholar]
- 199.Bartlett JB, Dredge K, and Dalgleish AG (2004). The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat Rev Cancer 4, 314–322. [DOI] [PubMed] [Google Scholar]
- 200.Sampaio EP, Sarno EN, Galilly R, Cohn ZA, and Kaplan G (1991). Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes. The Journal of experimental medicine 173, 699–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Moreira AL, Sampaio EP, Zmuidzinas A, Frindt P, Smith KA, and Kaplan G (1993). Thalidomide exerts its inhibitory action on tumor necrosis factor alpha by enhancing mRNA degradation. The Journal of experimental medicine 177, 1675–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.D’Amato RJ, Loughnan MS, Flynn E, and Folkman J (1994). Thalidomide is an inhibitor of angiogenesis. Proc Natl Acad Sci U S A 91, 4082–4085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Eddlemon P, Munshi N, Anaissie E, Wilson C, Dhodapkar M, et al. (1999). Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med 341, 1565–1571. [DOI] [PubMed] [Google Scholar]
- 204.Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau JL, Dmoszynska A, San Miguel J, Hellmann A, Facon T, Foa R, et al. (2007). Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 357, 2123–2132. [DOI] [PubMed] [Google Scholar]
- 205.Miguel JS, Weisel K, Moreau P, Lacy M, Song K, Delforge M, Karlin L, Goldschmidt H, Banos A, Oriol A, et al. (2013). Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, phase 3 trial. Lancet Oncol 14, 1055–1066. [DOI] [PubMed] [Google Scholar]
- 206.Muller GW, Corral LG, Shire MG, Wang H, Moreira A, Kaplan G, and Stirling DI (1996). Structural modifications of thalidomide produce analogs with enhanced tumor necrosis factor inhibitory activity. J Med Chem 39, 3238–3240. [DOI] [PubMed] [Google Scholar]
- 207.Schey SA, Fields P, Bartlett JB, Clarke IA, Ashan G, Knight RD, Streetly M, and Dalgleish AG (2004). Phase I study of an immunomodulatory thalidomide analog, CC-4047, in relapsed or refractory multiple myeloma. J Clin Oncol 22, 3269–3276. [DOI] [PubMed] [Google Scholar]
- 208.Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, Siegel D, Borrello I, Rajkumar SV, Chanan-Khan AA, et al. (2007). Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 357, 2133–2142. [DOI] [PubMed] [Google Scholar]
- 209.Habermann TM, Lossos IS, Justice G, Vose JM, Wiernik PH, McBride K, Wride K, Ervin-Haynes A, Takeshita K, Pietronigro D, et al. (2009). Lenalidomide oral monotherapy produces a high response rate in patients with relapsed or refractory mantle cell lymphoma. Br J Haematol 145, 344–349. [DOI] [PubMed] [Google Scholar]
- 210.List A, Kurtin S, Roe DJ, Buresh A, Mahadevan D, Fuchs D, Rimsza L, Heaton R, Knight R, and Zeldis JB (2005). Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med 352, 549–557. [DOI] [PubMed] [Google Scholar]
- 211.Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, Yamaguchi Y, and Handa H (2010). Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350. [DOI] [PubMed] [Google Scholar]
- 212.Fischer ES, Bohm K, Lydeard JR, Yang H, Stadler MB, Cavadini S, Nagel J, Serluca F, Acker V, Lingaraju GM, et al. (2014). Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 512, 49–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Kronke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, Svinkina T, Heckl D, Comer E, Li X, et al. (2014). Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Lu G, Middleton RE, Sun H, Naniong M, Ott CJ, Mitsiades CS, Wong KK, Bradner JE, and Kaelin WG Jr. (2014). The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 343, 305–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Winter GE, Buckley DL, Paulk J, Roberts JM, Souza A, Dhe-Paganon S, and Bradner JE (2015). DRUG DEVELOPMENT. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, and Deshaies RJ (2001). Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc Natl Acad Sci U S A 98, 8554–8559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Pollack VA, Savage DM, Baker DA, Tsaparikos KE, Sloan DE, Moyer JD, Barbacci EG, Pustilnik LR, Smolarek TA, Davis JA, et al. (1999). Inhibition of epidermal growth factor receptor-associated tyrosine phosphorylation in human carcinomas with CP-358,774: dynamics of receptor inhibition in situ and antitumor effects in athymic mice. J Pharmacol Exp Ther 291, 739–748. [PubMed] [Google Scholar]
- 218.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, et al. (2004). Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350, 2129–2139. [DOI] [PubMed] [Google Scholar]
- 219.Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, Singh B, Heelan R, Rusch V, Fulton L, et al. (2004). EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A 101, 13306–13311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Cohen MH, Johnson JR, Chen YF, Sridhara R, and Pazdur R (2005). FDA drug approval summary: erlotinib (Tarceva) tablets. Oncologist 10, 461–466. [DOI] [PubMed] [Google Scholar]
- 221.Li Z, Xu M, Xing S, Ho WT, Ishii T, Li Q, Fu X, and Zhao ZJ (2007). Erlotinib effectively inhibits JAK2V617F activity and polycythemia vera cell growth. J Biol Chem 282, 3428–3432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Boehrer S, Ades L, Braun T, Galluzzi L, Grosjean J, Fabre C, Le Roux G, Gardin C, Martin A, de Botton S, et al. (2008). Erlotinib exhibits antineoplastic off-target effects in AML and MDS: a preclinical study. Blood 111, 2170–2180. [DOI] [PubMed] [Google Scholar]
- 223.Weber C, Schreiber TB, and Daub H (2012). Dual phosphoproteomics and chemical proteomics analysis of erlotinib and gefitinib interference in acute myeloid leukemia cells. J Proteomics 75, 1343–1356. [DOI] [PubMed] [Google Scholar]
- 224.Thepot S, Boehrer S, Seegers V, Prebet T, Beyne-Rauzy O, Wattel E, Delaunay J, Raffoux E, Hunault M, Jourdan E, et al. (2014). A phase I/II trial of Erlotinib in higher risk myelodysplastic syndromes and acute myeloid leukemia after azacitidine failure. Leuk Res 38, 1430–1434. [DOI] [PubMed] [Google Scholar]
- 225.Komrokji RS, Padron E, Yu D, Fulp WJ, Rodriguez Y, Tinsley S, List AF, and Lancet JE (2014). Phase II clinical study of erlotinib for treatment of myelodysplastic syndromes. Am J Hematol 89, 809–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Abou Dalle I, Cortes JE, Pinnamaneni P, Lamothe B, Diaz Duque A, Randhawa J, Pemmaraju N, Jabbour E, Ferrajoli A, Wierda WG, et al. (2018). A Pilot Phase II Study of Erlotinib for the Treatment of Patients with Relapsed/Refractory Acute Myeloid Leukemia. Acta Haematol 140, 30–39. [DOI] [PubMed] [Google Scholar]
- 227.Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C, Faderl S, Ferrajoli A, Borthakur G, George S, et al. (2012). Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood 119, 4614–4618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Harrington EA, Bebbington D, Moore J, Rasmussen RK, Ajose-Adeogun AO, Nakayama T, Graham JA, Demur C, Hercend T, Diu-Hercend A, et al. (2004). VX-680, a potent and selective small-molecule inhibitor of the Aurora kinases, suppresses tumor growth in vivo. Nat Med 10, 262–267. [DOI] [PubMed] [Google Scholar]
- 229.Carter TA, Wodicka LM, Shah NP, Velasco AM, Fabian MA, Treiber DK, Milanov ZV, Atteridge CE, Biggs WH 3rd, Edeen PT, et al. (2005). Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases. Proc Natl Acad Sci U S A 102, 11011–11016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Young MA, Shah NP, Chao LH, Seeliger M, Milanov ZV, Biggs WH 3rd, Treiber DK, Patel HK, Zarrinkar PP, Lockhart DJ, et al. (2006). Structure of the kinase domain of an imatinib-resistant Abl mutant in complex with the Aurora kinase inhibitor VX-680. Cancer Res 66, 1007–1014. [DOI] [PubMed] [Google Scholar]
- 231.Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J, and Sawyers CL (2002). Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell 2, 117–125. [DOI] [PubMed] [Google Scholar]
- 232.Giles FJ, Cortes J, Jones D, Bergstrom D, Kantarjian H, and Freedman SJ (2007). MK-0457, a novel kinase inhibitor, is active in patients with chronic myeloid leukemia or acute lymphocytic leukemia with the T315I BCR-ABL mutation. Blood 109, 500–502. [DOI] [PubMed] [Google Scholar]
- 233.Sanchez-Guijo FM, Lopez-Jimenez J, Gonzalez T, Santamaria C, Gonzalez M, and Del Canizo MC (2009). Multitargeted sequential therapy with MK-0457 and dasatinib followed by stem cell transplantation for T315I mutated chronic myeloid leukemia. Leuk Res 33, e20–22. [DOI] [PubMed] [Google Scholar]
- 234.Giles FJ, Swords RT, Nagler A, Hochhaus A, Ottmann OG, Rizzieri DA, Talpaz M, Clark J, Watson P, Xiao A, et al. (2013). MK-0457, an Aurora kinase and BCR-ABL inhibitor, is active in patients with BCR-ABL T315I leukemia. Leukemia 27, 113–117. [DOI] [PubMed] [Google Scholar]
- 235.Seymour JF, Kim DW, Rubin E, Haregewoin A, Clark J, Watson P, Hughes T, Dufva I, Jimenez JL, Mahon FX, et al. (2014). A phase 2 study of MK-0457 in patients with BCR-ABL T315I mutant chronic myelogenous leukemia and philadelphia chromosome-positive acute lymphoblastic leukemia. Blood Cancer J 4, e238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Brell JM (2010). Prolonged QTc interval in cancer therapeutic drug development: defining arrhythmic risk in malignancy. Prog Cardiovasc Dis 53, 164–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Gillman PK (2007). Tricyclic antidepressant pharmacology and therapeutic drug interactions updated. Br J Pharmacol 151, 737–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Jeon SH, Kim SH, Kim Y, Kim YS, Lim Y, Lee YH, and Shin SY (2011). The tricyclic antidepressant imipramine induces autophagic cell death in U-87MG glioma cells. Biochem Biophys Res Commun 413, 311–317. [DOI] [PubMed] [Google Scholar]
- 239.Kabolizadeh P, Engelmann BJ, Pullen N, Stewart JK, Ryan JJ, and Farrell NP (2012). Platinum anticancer agents and antidepressants: desipramine enhances platinum-based cytotoxicity in human colon cancer cells. J Biol Inorg Chem 17, 123–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Jahchan NS, Dudley JT, Mazur PK, Flores N, Yang D, Palmerton A, Zmoos AF, Vaka D, Tran KQ, Zhou M, et al. (2013). A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discov 3, 1364–1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K, Castaneda S, Cornelius LA, Das J, Doweyko AM, et al. (2004). Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J Med Chem 47, 6658–6661. [DOI] [PubMed] [Google Scholar]
- 242.Haura EB, Tanvetyanon T, Chiappori A, Williams C, Simon G, Antonia S, Gray J, Litschauer S, Tetteh L, Neuger A, et al. (2010). Phase I/II study of the Src inhibitor dasatinib in combination with erlotinib in advanced non-small-cell lung cancer. J Clin Oncol 28, 1387–1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Li J, Rix U, Fang B, Bai Y, Edwards A, Colinge J, Bennett KL, Gao J, Song L, Eschrich S, et al. (2010). A chemical and phosphoproteomic characterization of dasatinib action in lung cancer. Nat Chem Biol 6, 291–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Song L, Morris M, Bagui T, Lee FY, Jove R, and Haura EB (2006). Dasatinib (BMS-354825) selectively induces apoptosis in lung cancer cells dependent on epidermal growth factor receptor signaling for survival. Cancer Res 66, 5542–5548. [DOI] [PubMed] [Google Scholar]
- 245.Hammerman PS, Sos ML, Ramos AH, Xu C, Dutt A, Zhou W, Brace LE, Woods BA, Lin W, Zhang J, et al. (2011). Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer discovery 1, 78–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Day E, Waters B, Spiegel K, Alnadaf T, Manley PW, Buchdunger E, Walker C, and Jarai G (2008). Inhibition of collagen-induced discoidin domain receptor 1 and 2 activation by imatinib, nilotinib and dasatinib. Eur J Pharmacol 599, 44–53. [DOI] [PubMed] [Google Scholar]
- 247.Rix U, Hantschel O, Durnberger G, Remsing Rix LL, Planyavsky M, Fernbach NV, Kaupe I, Bennett KL, Valent P, Colinge J, et al. (2007). Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 110, 4055–4063. [DOI] [PubMed] [Google Scholar]
- 248.Hantschel O, Rix U, Schmidt U, Burckstummer T, Kneidinger M, Schutze G, Colinge J, Bennett KL, Ellmeier W, Valent P, et al. (2007). The Btk tyrosine kinase is a major target of the Bcr-Abl inhibitor dasatinib. Proc Natl Acad Sci U S A 104, 13283–13288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Sillaber C, Herrmann H, Bennett K, Rix U, Baumgartner C, Bohm A, Herndlhofer S, Tschachler E, Superti-Furga G, Jager U, et al. (2009). Immunosuppression and atypical infections in CML patients treated with dasatinib at 140 mg daily. Eur J Clin Invest 39, 1098–1109. [DOI] [PubMed] [Google Scholar]
- 250.Bai Y, Kim JY, Watters JM, Fang B, Kinose F, Song L, Koomen JM, Teer JK, Fisher K, Chen YA, et al. (2014). Adaptive responses to dasatinib-treated lung squamous cell cancer cells harboring DDR2 mutations. Cancer Res 74, 7217–7228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Liu L, Hussain M, Luo J, Duan A, Chen C, Tu Z, and Zhang J (2017). Synthesis and biological evaluation of novel dasatinib analogues as potent DDR1 and DDR2 kinase inhibitors. Chemical biology & drug design 89, 420–427. [DOI] [PubMed] [Google Scholar]
- 252.Terai H, Tan L, Beauchamp EM, Hatcher JM, Liu Q, Meyerson M, Gray NS, and Hammerman PS (2015). Characterization of DDR2 Inhibitors for the Treatment of DDR2 Mutated Nonsmall Cell Lung Cancer. ACS chemical biology 10, 2687–2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Brown JR (2013). Ibrutinib (PCI-32765), the first BTK (Bruton’s tyrosine kinase) inhibitor in clinical trials. Curr Hematol Malig Rep 8, 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B, Li S, Pan Z, Thamm DH, Miller RA, et al. (2010). The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci U S A 107, 13075–13080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Pan Z, Scheerens H, Li SJ, Schultz BE, Sprengeler PA, Burrill LC, Mendonca RV, Sweeney MD, Scott KC, Grothaus PG, et al. (2007). Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. ChemMedChem 2, 58–61. [DOI] [PubMed] [Google Scholar]
- 256.Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, Kohlhammer H, Lamy L, Zhao H, Yang Y, et al. (2010). Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 463, 88–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Scuoppo C, Wang J, Persaud M, Mittan SK, Basso K, Pasqualucci L, Rabadan R, Inghirami G, Grandori C, Bosch F, et al. (2019). Repurposing dasatinib for diffuse large B cell lymphoma. Proc Natl Acad Sci U S A 116, 16981–16986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Yang C, Lu P, Lee FY, Chadburn A, Barrientos JC, Leonard JP, Ye F, Zhang D, Knowles DM, and Wang YL (2008). Tyrosine kinase inhibition in diffuse large B-cell lymphoma: molecular basis for antitumor activity and drug resistance of dasatinib. Leukemia 22, 1755–1766. [DOI] [PubMed] [Google Scholar]
- 259.Young RM, and Staudt LM (2014). Ibrutinib treatment of CLL: the cancer fights back. Cancer Cell 26, 11–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Sheridan C (2012). Companies in rapid pursuit of Btk immunokinase. Nat Biotechnol 30, 199–200. [DOI] [PubMed] [Google Scholar]
- 261.Gao W, Wang M, Wang L, Lu H, Wu S, Dai B, Ou Z, Zhang L, Heymach JV, Gold KA, et al. (2014). Selective antitumor activity of ibrutinib in EGFR-mutant non-small cell lung cancer cells. J Natl Cancer Inst 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Grabinski N, and Ewald F (2014). Ibrutinib (ImbruvicaTM) potently inhibits ErbB receptor phosphorylation and cell viability of ErbB2-positive breast cancer cells. Investigational new drugs 32, 1096–1104. [DOI] [PubMed] [Google Scholar]
- 263.Campbell R, Chong G, and Hawkes EA (2018). Novel Indications for Bruton’s Tyrosine Kinase Inhibitors, beyond Hematological Malignancies. J Clin Med 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Haura EB, and Rix U (2014). Deploying ibrutinib to lung cancer: another step in the quest towards drug repurposing. J Natl Cancer Inst 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Wang A, Yan XE, Wu H, Wang W, Hu C, Chen C, Zhao Z, Zhao P, Li X, Wang L, et al. (2016). Ibrutinib targets mutant-EGFR kinase with a distinct binding conformation. Oncotarget 7, 69760–69769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Wu H, Wang A, Zhang W, Wang B, Chen C, Wang W, Hu C, Ye Z, Zhao Z, Wang L, et al. (2015). Ibrutinib selectively and irreversibly targets EGFR (L858R, Del19) mutant but is moderately resistant to EGFR (T790M) mutant NSCLC Cells. Oncotarget 6, 31313–31322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Hu J, Zhang H, Cao M, Wang L, Wu S, and Fang B (2018). Auranofin Enhances Ibrutinib’s Anticancer Activity in EGFR-Mutant Lung Adenocarcinoma. Mol Cancer Ther 17, 2156–2163. [DOI] [PubMed] [Google Scholar]
- 268.Chen J, Kinoshita T, Sukbuntherng J, Chang BY, and Elias L (2016). Ibrutinib Inhibits ERBB Receptor Tyrosine Kinases and HER2-Amplified Breast Cancer Cell Growth. Mol Cancer Ther 15, 2835–2844. [DOI] [PubMed] [Google Scholar]
- 269.Masso-Valles D, Jauset T, Serrano E, Sodir NM, Pedersen K, Affara NI, Whitfield JR, Beaulieu ME, Evan GI, Elias L, et al. (2015). Ibrutinib exerts potent antifibrotic and antitumor activities in mouse models of pancreatic adenocarcinoma. Cancer Res 75, 1675–1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Stiff A, Trikha P, Wesolowski R, Kendra K, Hsu V, Uppati S, McMichael E, Duggan M, Campbell A, Keller K, et al. (2016). Myeloid-Derived Suppressor Cells Express Bruton’s Tyrosine Kinase and Can Be Depleted in Tumor-Bearing Hosts by Ibrutinib Treatment. Cancer Res 76, 2125–2136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Rauf F, Festa F, Park JG, Magee M, Eaton S, Rinaldi C, Betanzos CM, Gonzalez-Malerva L, and LaBaer J (2018). Ibrutinib inhibition of ERBB4 reduces cell growth in a WNT5A-dependent manner. Oncogene 37, 2237–2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Uitdehaag JCM, Kooijman JJ, de Roos J, Prinsen MBW, Dylus J, Willemsen-Seegers N, Kawase Y, Sawa M, de Man J, van Gerwen SJC, et al. (2019). Combined Cellular and Biochemical Profiling to Identify Predictive Drug Response Biomarkers for Kinase Inhibitors Approved for Clinical Use between 2013 and 2017. Mol Cancer Ther 18, 470–481. [DOI] [PubMed] [Google Scholar]
- 273.Stephens DM, and Byrd JC (2019). How I manage ibrutinib intolerance and complications in patients with chronic lymphocytic leukemia. Blood 133, 1298–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Inai T, Mancuso M, Hashizume H, Baffert F, Haskell A, Baluk P, Hu-Lowe DD, Shalinsky DR, Thurston G, Yancopoulos GD, et al. (2004). Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am J Pathol 165, 35–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG, et al. (2011). Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378, 1931–1939. [DOI] [PubMed] [Google Scholar]
- 276.Pemovska T, Johnson E, Kontro M, Repasky GA, Chen J, Wells P, Cronin CN, McTigue M, Kallioniemi O, Porkka K, et al. (2015). Axitinib effectively inhibits BCR-ABL1(T315I) with a distinct binding conformation. Nature 519, 102–105. [DOI] [PubMed] [Google Scholar]
- 277.Zabriskie MS, Eide CA, Yan D, Vellore NA, Pomicter AD, Savage SL, Druker BJ, Deininger MW, and O’Hare T (2016). Extreme mutational selectivity of axitinib limits its potential use as a targeted therapeutic for BCR-ABL1-positive leukemia. Leukemia 30, 1418–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Winter GE, Rix U, Carlson SM, Gleixner KV, Grebien F, Gridling M, Muller AC, Breitwieser FP, Bilban M, Colinge J, et al. (2012). Systems-pharmacology dissection of a drug synergy in imatinib-resistant CML. Nat Chem Biol 8, 905–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Huang WS, Metcalf CA, Sundaramoorthi R, Wang Y, Zou D, Thomas RM, Zhu X, Cai L, Wen D, Liu S, et al. (2010). Discovery of 3-[2-(imidazo[1,2-b]pyridazin-3-yl)ethynyl]-4-methyl-N-{4-[(4-methylpiperazin-1-y l)methyl]-3-(trifluoromethyl)phenyl}benzamide (AP24534), a potent, orally active pan-inhibitor of breakpoint cluster region-abelson (BCR-ABL) kinase including the T315I gatekeeper mutant. J Med Chem 53, 4701–4719. [DOI] [PubMed] [Google Scholar]
- 280.O’Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, Adrian LT, Zhou T, Huang WS, Xu Q, et al. (2009). AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 16, 401–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Gozgit JM, Wong MJ, Moran L, Wardwell S, Mohemmad QK, Narasimhan NI, Shakespeare WC, Wang F, Clackson T, and Rivera VM (2012). Ponatinib (AP24534), a multitargeted pan-FGFR inhibitor with activity in multiple FGFR-amplified or mutated cancer models. Mol Cancer Ther 11, 690–699. [DOI] [PubMed] [Google Scholar]
- 282.Gozgit JM, Wong MJ, Wardwell S, Tyner JW, Loriaux MM, Mohemmad QK, Narasimhan NI, Shakespeare WC, Wang F, Druker BJ, et al. (2011). Potent activity of ponatinib (AP24534) in models of FLT3-driven acute myeloid leukemia and other hematologic malignancies. Mol Cancer Ther 10, 1028–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Smith CC, Lasater EA, Zhu X, Lin KC, Stewart WK, Damon LE, Salerno S, and Shah NP (2013). Activity of ponatinib against clinically-relevant AC220-resistant kinase domain mutants of FLT3-ITD. Blood 121, 3165–3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, Harris PL, Driscoll DR, Fidias P, Lynch TJ, et al. (2005). Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci U S A 102, 7665–7670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Lu L, Ghose AK, Quail MR, Albom MS, Durkin JT, Holskin BP, Angeles TS, Meyer SL, Ruggeri BA, and Cheng M (2009). ALK mutants in the kinase domain exhibit altered kinase activity and differential sensitivity to small molecule ALK inhibitors. Biochemistry 48, 3600–3609. [DOI] [PubMed] [Google Scholar]
- 286.Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, Kris MG, and Varmus H (2005). Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2, e73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.De Falco V, Buonocore P, Muthu M, Torregrossa L, Basolo F, Billaud M, Gozgit JM, Carlomagno F, and Santoro M (2013). Ponatinib (AP24534) is a novel potent inhibitor of oncogenic RET mutants associated with thyroid cancer. The Journal of clinical endocrinology and metabolism 98, E811–819. [DOI] [PubMed] [Google Scholar]
- 288.Mologni L, Redaelli S, Morandi A, Plaza-Menacho I, and Gambacorti-Passerini C (2013). Ponatinib is a potent inhibitor of wild-type and drug-resistant gatekeeper mutant RET kinase. Mol Cell Endocrinol 377, 1–6. [DOI] [PubMed] [Google Scholar]
- 289.Arighi E, Borrello MG, and Sariola H (2005). RET tyrosine kinase signaling in development and cancer. Cytokine Growth Factor Rev 16, 441–467. [DOI] [PubMed] [Google Scholar]
- 290.Gozgit JM, Chen TH, Song Y, Wardwell S, Wang F, Cai J, Li H, Edgren H, Rivera VM, and Pritchard J (2018). RET fusions observed in lung and colorectal cancers are sensitive to ponatinib. Oncotarget 9, 29654–29664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Tan FH, Putoczki TL, Stylli SS, and Luwor RB (2019). Ponatinib: a novel multi-tyrosine kinase inhibitor against human malignancies. Onco Targets Ther 12, 635–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Tannir NM, Schwab G, and Grunwald V (2017). Cabozantinib: an Active Novel Multikinase Inhibitor in Renal Cell Carcinoma. Current oncology reports 19, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.You WK, Sennino B, Williamson CW, Falcon B, Hashizume H, Yao LC, Aftab DT, and McDonald DM (2011). VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer. Cancer Res 71, 4758–4768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Chong CR, Bahcall M, Capelletti M, Kosaka T, Ercan D, Sim T, Sholl LM, Nishino M, Johnson BE, Gray NS, et al. (2017). Identification of Existing Drugs That Effectively Target NTRK1 and ROS1 Rearrangements in Lung Cancer. Clin Cancer Res 23, 204–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Davare MA, Vellore NA, Wagner JP, Eide CA, Goodman JR, Drilon A, Deininger MW, O’Hare T, and Druker BJ (2015). Structural insight into selectivity and resistance profiles of ROS1 tyrosine kinase inhibitors. Proc Natl Acad Sci U S A 112, E5381–5390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Fuse MJ, Okada K, Oh-Hara T, Ogura H, Fujita N, and Katayama R (2017). Mechanisms of Resistance to NTRK Inhibitors and Therapeutic Strategies in NTRK1-Rearranged Cancers. Mol Cancer Ther 16, 2130–2143. [DOI] [PubMed] [Google Scholar]
- 297.Katayama R, Kobayashi Y, Friboulet L, Lockerman EL, Koike S, Shaw AT, Engelman JA, and Fujita N (2015). Cabozantinib overcomes crizotinib resistance in ROS1 fusion-positive cancer. Clin Cancer Res 21, 166–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Bongarzone I, Pierotti MA, Monzini N, Mondellini P, Manenti G, Donghi R, Pilotti S, Grieco M, Santoro M, Fusco A, et al. (1989). High frequency of activation of tyrosine kinase oncogenes in human papillary thyroid carcinoma. Oncogene 4, 1457–1462. [PubMed] [Google Scholar]
- 299.Martin-Zanca D, Hughes SH, and Barbacid M (1986). A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature 319, 743–748. [DOI] [PubMed] [Google Scholar]
- 300.Cocco E, Scaltriti M, and Drilon A (2018). NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 15, 731–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Drilon A, Somwar R, Wagner JP, Vellore NA, Eide CA, Zabriskie MS, Arcila ME, Hechtman JF, Wang L, Smith RS, et al. (2016). A Novel Crizotinib-Resistant Solvent-Front Mutation Responsive to Cabozantinib Therapy in a Patient with ROS1-Rearranged Lung Cancer. Clin Cancer Res 22, 2351–2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Sun TY, Niu X, Chakraborty A, Neal JW, and Wakelee HA (2019). Lengthy Progression-Free Survival and Intracranial Activity of Cabozantinib in Patients with Crizotinib and Ceritinib-Resistant ROS1- Positive Non-Small Cell Lung Cancer. J Thorac Oncol 14, e21–e24. [DOI] [PubMed] [Google Scholar]
- 303.Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, Michellys PY, Awad MM, Yanagitani N, Kim S, et al. (2014). The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov 4, 662–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Gainor JF, Tan DS, De Pas T, Solomon BJ, Ahmad A, Lazzari C, de Marinis F, Spitaleri G, Schultz K, Friboulet L, et al. (2015). Progression-Free and Overall Survival in ALK-Positive NSCLC Patients Treated with Sequential Crizotinib and Ceritinib. Clin Cancer Res 21, 2745–2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Marsilje TH, Pei W, Chen B, Lu W, Uno T, Jin Y, Jiang T, Kim S, Li N, Warmuth M, et al. (2013). Synthesis, structure-activity relationships, and in vivo efficacy of the novel potent and selective anaplastic lymphoma kinase (ALK) inhibitor 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulf onyl)phenyl)pyrimidine-2,4-diamine (LDK378) currently in phase 1 and phase 2 clinical trials. J Med Chem 56, 5675–5690. [DOI] [PubMed] [Google Scholar]
- 306.Facchinetti F, Loriot Y, Kuo MS, Mahjoubi L, Lacroix L, Planchard D, Besse B, Farace F, Auger N, Remon J, et al. (2016). Crizotinib-Resistant ROS1 Mutations Reveal a Predictive Kinase Inhibitor Sensitivity Model for ROS1- and ALK-Rearranged Lung Cancers. Clin Cancer Res 22, 5983–5991. [DOI] [PubMed] [Google Scholar]
- 307.Lim SM, Kim HR, Lee JS, Lee KH, Lee YG, Min YJ, Cho EK, Lee SS, Kim BS, Choi MY, et al. (2017). Open-Label, Multicenter, Phase II Study of Ceritinib in Patients With Non-Small-Cell Lung Cancer Harboring ROS1 Rearrangement. J Clin Oncol 35, 2613–2618. [DOI] [PubMed] [Google Scholar]
- 308.Lovly CM, McDonald NT, Chen H, Ortiz-Cuaran S, Heukamp LC, Yan Y, Florin A, Ozretic L, Lim D, Wang L, et al. (2014). Rationale for co-targeting IGF-1R and ALK in ALK fusion-positive lung cancer. Nat Med 20, 1027–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.van Erp AEM, Hillebrandt-Roeffen MHS, van Houdt L, Fleuren EDG, van der Graaf WTA, and Versleijen-Jonkers YMH (2017). Targeting Anaplastic Lymphoma Kinase (ALK) in Rhabdomyosarcoma (RMS) with the Second-Generation ALK Inhibitor Ceritinib. Target Oncol 12, 815–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Wang F, Bank T, Malnassy G, Arteaga M, Shang N, Dalheim A, Ding X, Cotler SJ, Denning MF, Nishimura MI, et al. (2018). Inhibition of insulin-like growth factor 1 receptor enhances the efficacy of sorafenib in inhibiting hepatocellular carcinoma cell growth and survival. Hepatol Commun 2, 732–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Verduzco D, Kuenzi BM, Kinose F, Sondak VK, Eroglu Z, Rix U, and Smalley KSM (2018). Ceritinib Enhances the Efficacy of Trametinib in BRAF/NRAS-Wild-Type Melanoma Cell Lines. Mol Cancer Ther 17, 73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Pantziarka P, Sukhatme V, Bouche G, Meheus L, and Sukhatme VP (2015). Repurposing Drugs in Oncology (ReDO)-itraconazole as an anti-cancer agent. Ecancermedicalscience 9, 521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313.Vanden Bossche H, Marichal P, Le Jeune L, Coene MC, Gorrens J, and Cools W (1993). Effects of itraconazole on cytochrome P-450-dependent sterol 14 alpha-demethylation and reduction of 3-ketosteroids in Cryptococcus neoformans. Antimicrob Agents Chemother 37, 2101–2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Chong CR, Xu J, Lu J, Bhat S, Sullivan DJ Jr., and Liu JO (2007). Inhibition of angiogenesis by the antifungal drug itraconazole. ACS chemical biology 2, 263–270. [DOI] [PubMed] [Google Scholar]
- 315.Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS, Fereshteh M, Gardner D, et al. (2010). Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell 17, 388–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Aftab BT, Dobromilskaya I, Liu JO, and Rudin CM (2011). Itraconazole inhibits angiogenesis and tumor growth in non-small cell lung cancer. Cancer Res 71, 6764–6772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Kim J, Aftab BT, Tang JY, Kim D, Lee AH, Rezaee M, Kim J, Chen B, King EM, Borodovsky A, et al. (2013). Itraconazole and arsenic trioxide inhibit Hedgehog pathway activation and tumor growth associated with acquired resistance to smoothened antagonists. Cancer Cell 23, 23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.Tsai YC, and Tsai TF (2019). Itraconazole in the Treatment of Nonfungal Cutaneous Diseases: A Review. Dermatol Ther (Heidelb) 9, 271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Tsubamoto H, Ueda T, Inoue K, Sakata K, Shibahara H, and Sonoda T (2017). Repurposing itraconazole as an anticancer agent. Oncol Lett 14, 1240–1246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Kim DJ, Kim J, Spaunhurst K, Montoya J, Khodosh R, Chandra K, Fu T, Gilliam A, Molgo M, Beachy PA, et al. (2014). Open-label, exploratory phase II trial of oral itraconazole for the treatment of basal cell carcinoma. J Clin Oncol 32, 745–751. [DOI] [PubMed] [Google Scholar]
- 321.Fung SL, Chau CH, and Yew WW (2008). Cardiovascular adverse effects during itraconazole therapy. Eur Respir J 32, 240. [DOI] [PubMed] [Google Scholar]
- 322.Vollenbroich R, Maeder MT, and Weilenmann D (2014). Congestive heart failure related to antifungal therapy with itraconazole. Int J Cardiol 172, e170–171. [DOI] [PubMed] [Google Scholar]
- 323.Viola-Rhenals M, Patel KR, Jaimes-Santamaria L, Wu G, Liu J, and Dou QP (2018). Recent Advances in Antabuse (Disulfiram): The Importance of its Metal-binding Ability to its Anticancer Activity. Current medicinal chemistry 25, 506–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Lovborg H, Oberg F, Rickardson L, Gullbo J, Nygren P, and Larsson R (2006). Inhibition of proteasome activity, nuclear factor-KappaB translocation and cell survival by the antialcoholism drug disulfiram. Int J Cancer 118, 1577–1580. [DOI] [PubMed] [Google Scholar]
- 325.Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, Ozdian T, Bartkova J, Turi Z, Moudry P, et al. (2017). Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature 552, 194–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Rout S, and Mahapatra RK (2019). Plasmodium falciparum: Multidrug resistance. Chemical biology & drug design 93, 737–759. [DOI] [PubMed] [Google Scholar]
- 327.Verbaanderd C, Maes H, Schaaf MB, Sukhatme VP, Pantziarka P, Sukhatme V, Agostinis P, and Bouche G (2017). Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience 11, 781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.Rebecca VW, Nicastri MC, Fennelly C, Chude CI, Barber-Rotenberg JS, Ronghe A, McAfee Q, McLaughlin NP, Zhang G, Goldman AR, et al. (2019). PPT1 Promotes Tumor Growth and Is the Molecular Target of Chloroquine Derivatives in Cancer. Cancer Discov 9, 220–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329.Rebecca VW, Nicastri MC, McLaughlin N, Fennelly C, McAfee Q, Ronghe A, Nofal M, Lim CY, Witze E, Chude CI, et al. (2017). A Unified Approach to Targeting the Lysosome’s Degradative and Growth Signaling Roles. Cancer Discov 7, 1266–1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.McAfee Q, Zhang Z, Samanta A, Levi SM, Ma XH, Piao S, Lynch JP, Uehara T, Sepulveda AR, Davis LE, et al. (2012). Autophagy inhibitor Lys05 has single-agent antitumor activity and reproduces the phenotype of a genetic autophagy deficiency. Proc Natl Acad Sci U S A 109, 8253–8258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331.Bhaw-Luximon A, and Jhurry D (2017). Artemisinin and its derivatives in cancer therapy: status of progress, mechanism of action, and future perspectives. Cancer Chemother Pharmacol 79, 451–466. [DOI] [PubMed] [Google Scholar]
- 332.Tu Y (2011). The discovery of artemisinin (qinghaosu) and gifts from Chinese medicine. Nat Med 17, 1217–1220. [DOI] [PubMed] [Google Scholar]
- 333.O’Neill PM, Barton VE, and Ward SA (2010). The molecular mechanism of action of artemisinin--the debate continues. Molecules 15, 1705–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334.Gotsbacher MP, Cho SM, Kim NH, Liu F, Kwon HJ, and Karuso P (2019). Reverse Chemical Proteomics Identifies an Unanticipated Human Target of the Antimalarial Artesunate. ACS chemical biology 14, 636–643. [DOI] [PubMed] [Google Scholar]
- 335.Chidley C, Haruki H, Pedersen MG, Muller E, and Johnsson K (2011). A yeast-based screen reveals that sulfasalazine inhibits tetrahydrobiopterin biosynthesis. Nat Chem Biol 7, 375–383. [DOI] [PubMed] [Google Scholar]
- 336.Yang S, Jan YH, Mishin V, Richardson JR, Hossain MM, Heindel ND, Heck DE, Laskin DL, and Laskin JD (2015). Sulfa drugs inhibit sepiapterin reduction and chemical redox cycling by sepiapterin reductase. J Pharmacol Exp Ther 352, 529–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Costigan M, Latremoliere A, and Woolf CJ (2012). Analgesia by inhibiting tetrahydrobiopterin synthesis. Curr Opin Pharmacol 12, 92–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338.McNeill E, and Channon KM (2012). The role of tetrahydrobiopterin in inflammation and cardiovascular disease. Thromb Haemost 108, 832–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339.Mooney MR, Geerts D, Kort EJ, and Bachmann AS (2019). Anti-tumor effect of sulfasalazine in neuroblastoma. Biochem Pharmacol 162, 237–249. [DOI] [PubMed] [Google Scholar]
- 340.Hoelder S, Clarke PA, and Workman P (2012). Discovery of small molecule cancer drugs: successes, challenges and opportunities. Molecular oncology 6, 155–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341.Shah NP, Tran C, Lee FY, Chen P, Norris D, and Sawyers CL (2004). Overriding imatinib resistance with a novel ABL kinase inhibitor. Science 305, 399–401. [DOI] [PubMed] [Google Scholar]
- 342.Danhauser-Riedl S, Warmuth M, Druker BJ, Emmerich B, and Hallek M (1996). Activation of Src kinases p53/56lyn and p59hck by p210bcr/abl in myeloid cells. Cancer Res 56, 3589–3596. [PubMed] [Google Scholar]
- 343.Donato NJ, Wu JY, Stapley J, Gallick G, Lin H, Arlinghaus R, and Talpaz M (2003). BCR-ABL independence and LYN kinase overexpression in chronic myelogenous leukemia cells selected for resistance to STI571. Blood 101, 690–698. [DOI] [PubMed] [Google Scholar]
- 344.Hu Y, Liu Y, Pelletier S, Buchdunger E, Warmuth M, Fabbro D, Hallek M, Van Etten RA, and Li S (2004). Requirement of Src kinases Lyn, Hck and Fgr for BCR-ABL1-induced B-lymphoblastic leukemia but not chronic myeloid leukemia. Nature genetics 36, 453–461. [DOI] [PubMed] [Google Scholar]
- 345.Boschelli DH, Ye F, Wang YD, Dutia M, Johnson SL, Wu B, Miller K, Powell DW, Yaczko D, Young M, et al. (2001). Optimization of 4-phenylamino-3-quinolinecarbonitriles as potent inhibitors of Src kinase activity. J Med Chem 44, 3965–3977. [DOI] [PubMed] [Google Scholar]
- 346.Golas JM, Arndt K, Etienne C, Lucas J, Nardin D, Gibbons J, Frost P, Ye F, Boschelli DH, and Boschelli F (2003). SKI-606, a 4-anilino-3-quinolinecarbonitrile dual inhibitor of Src and Abl kinases, is a potent antiproliferative agent against chronic myelogenous leukemia cells in culture and causes regression of K562 xenografts in nude mice. Cancer Res 63, 375–381. [PubMed] [Google Scholar]
- 347.Kimura S, Naito H, Segawa H, Kuroda J, Yuasa T, Sato K, Yokota A, Kamitsuji Y, Kawata E, Ashihara E, et al. (2005). NS-187, a potent and selective dual Bcr-Abl/Lyn tyrosine kinase inhibitor, is a novel agent for imatinib-resistant leukemia. Blood 106, 3948–3954. [DOI] [PubMed] [Google Scholar]
- 348.Laird AD, Christensen JG, Li G, Carver J, Smith K, Xin X, Moss KG, Louie SG, Mendel DB, and Cherrington JM (2002). SU6668 inhibits Flk-1/KDR and PDGFRbeta in vivo, resulting in rapid apoptosis of tumor vasculature and tumor regression in mice. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 16, 681–690. [DOI] [PubMed] [Google Scholar]
- 349.Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, Schwartz B, Simantov R, and Kelley S (2006). Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov 5, 835–844. [DOI] [PubMed] [Google Scholar]
- 350.Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M, et al. (2004). BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 64, 7099–7109. [DOI] [PubMed] [Google Scholar]
- 351.Apsel B, Blair JA, Gonzalez B, Nazif TM, Feldman ME, Aizenstein B, Hoffman R, Williams RL, Shokat KM, and Knight ZA (2008). Targeted polypharmacology: discovery of dual inhibitors of tyrosine and phosphoinositide kinases. Nat Chem Biol 4, 691–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352.Cai X, Zhai HX, Wang J, Forrester J, Qu H, Yin L, Lai CJ, Bao R, and Qian C (2010). Discovery of 7-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yloxy)-N-hydroxyheptanamide (CUDc-101) as a potent multi-acting HDAC, EGFR, and HER2 inhibitor for the treatment of cancer. J Med Chem 53, 2000–2009. [DOI] [PubMed] [Google Scholar]
- 353.Lai CJ, Bao R, Tao X, Wang J, Atoyan R, Qu H, Wang DG, Yin L, Samson M, Forrester J, et al. (2010). CUDC-101, a multitargeted inhibitor of histone deacetylase, epidermal growth factor receptor, and human epidermal growth factor receptor 2, exerts potent anticancer activity. Cancer Res 70, 3647–3656. [DOI] [PubMed] [Google Scholar]
- 354.Qian C, Lai CJ, Bao R, Wang DG, Wang J, Xu GX, Atoyan R, Qu H, Yin L, Samson M, et al. (2012). Cancer network disruption by a single molecule inhibitor targeting both histone deacetylase activity and phosphatidylinositol 3-kinase signaling. Clin Cancer Res 18, 4104–4113. [DOI] [PubMed] [Google Scholar]
- 355.Yao L, Mustafa N, Tan EC, Poulsen A, Singh P, Duong-Thi MD, Lee JXT, Ramanujulu PM, Chng WJ, Yen JJY, et al. (2017). Design and Synthesis of Ligand Efficient Dual Inhibitors of Janus Kinase (JAK) and Histone Deacetylase (HDAC) Based on Ruxolitinib and Vorinostat. J Med Chem 60, 8336–8357. [DOI] [PubMed] [Google Scholar]
- 356.Liao Y, Niu X, Chen B, Edwards H, Xu L, Xie C, Lin H, Polin L, Taub JW, Ge Y, et al. (2016). Synthesis and Antileukemic Activities of Piperlongumine and HDAC Inhibitor Hybrids against Acute Myeloid Leukemia Cells. J Med Chem 59, 7974–7990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 357.He S, Dong G, Wang Z, Chen W, Huang Y, Li Z, Jiang Y, Liu N, Yao J, Miao Z, et al. (2015). Discovery of Novel Multiacting Topoisomerase I/II and Histone Deacetylase Inhibitors. ACS Med Chem Lett 6, 239–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358.Amemiya S, Yamaguchi T, Hashimoto Y, and Noguchi-Yachide T (2017). Synthesis and evaluation of novel dual BRD4/HDAC inhibitors. Bioorg Med Chem 25, 3677–3684. [DOI] [PubMed] [Google Scholar]
- 359.Ciceri P, Muller S, O’Mahony A, Fedorov O, Filippakopoulos P, Hunt JP, Lasater EA, Pallares G, Picaud S, Wells C, et al. (2014). Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat Chem Biol 10, 305–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360.Ember SW, Zhu JY, Olesen SH, Martin MP, Becker A, Berndt N, Georg GI, and Schonbrunn E (2014). Acetyl-lysine binding site of bromodomain-containing protein 4 (BRD4) interacts with diverse kinase inhibitors. ACS chemical biology 9, 1160–1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361.Ember SW, Lambert QT, Berndt N, Gunawan S, Ayaz M, Tauro M, Zhu JY, Cranfill PJ, Greninger P, Lynch CC, et al. (2017). Potent Dual BET Bromodomain-Kinase Inhibitors as Value-Added Multitargeted Chemical Probes and Cancer Therapeutics. Mol Cancer Ther 16, 1054–1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362.Qian F, Engst S, Yamaguchi K, Yu P, Won KA, Mock L, Lou T, Tan J, Li C, Tam D, et al. (2009). Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Res 69, 8009–8016. [DOI] [PubMed] [Google Scholar]
- 363.Sos ML, Dietlein F, Peifer M, Schottle J, Balke-Want H, Muller C, Koker M, Richters A, Heynck S, Malchers F, et al. (2012). A framework for identification of actionable cancer genome dependencies in small cell lung cancer. Proc Natl Acad Sci U S A 109, 17034–17039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364.Al-Ali H, Lee DH, Danzi MC, Nassif H, Gautam P, Wennerberg K, Zuercher B, Drewry DH, Lee JK, Lemmon VP, et al. (2015). Rational Polypharmacology: Systematically Identifying and Engaging Multiple Drug Targets To Promote Axon Growth. ACS chemical biology 10, 1939–1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 365.Gautam P, Jaiswal A, Aittokallio T, Al-Ali H, and Wennerberg K (2019). Phenotypic Screening Combined with Machine Learning for Efficient Identification of Breast Cancer-Selective Therapeutic Targets. Cell chemical biology 26, 970–979 e974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366.Andronis C, Sharma A, Virvilis V, Deftereos S, and Persidis A (2011). Literature mining, ontologies and information visualization for drug repurposing. Brief Bioinform 12, 357–368. [DOI] [PubMed] [Google Scholar]
- 367.Deftereos SN, Andronis C, Friedla EJ, Persidis A, and Persidis A (2011). Drug repurposing and adverse event prediction using high-throughput literature analysis. Wiley Interdiscip Rev Syst Biol Med 3, 323–334. [DOI] [PubMed] [Google Scholar]
- 368.Lim H, Poleksic A, Yao Y, Tong H, He D, Zhuang L, Meng P, and Xie L (2016). Large-Scale Off-Target Identification Using Fast and Accurate Dual Regularized One-Class Collaborative Filtering and Its Application to Drug Repurposing. PLoS Comput Biol 12, e1005135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369.Sanchez CG, Molinski SV, Gongora R, Sosulski M, Fuselier T, MacKinnon SS, Mondal D, and Lasky JA (2018). The Antiretroviral Agent Nelfinavir Mesylate: A Potential Therapy for Systemic Sclerosis. Arthritis Rheumatol 70, 115–126. [DOI] [PubMed] [Google Scholar]
