Skip to main content

Old drugs, new challenges: reassigning drugs for cancer therapies

Abstract

The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug’s approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs’ potential for new cancer therapies.

Introduction

Despite decades of ongoing efforts in the war against cancer, current therapeutic options often remain insufficient [1, 2]. The unresolved challenge is the complexity and intrinsic polygenicity of neoplastic diseases, which cannot be addressed by targeting a single molecular target. A combination of different treatment modalities is usually required to achieve optimal outcomes that often depend on the individual patient and cancer type. The development of effective and sophisticated treatment strategies is currently fueled by rapid progress in basic biomedical research as well as the evolution of surgical, radiotherapeutic, and immunological approaches.

Nevertheless, the safe application of antitumor drugs is limited by the off-target effects of most current drug strategies. Additionally, their prolonged use can result in modified tumor occurrences and resistance, as well as their ability to promote cancer stem cells [3]. Furthermore, with the acknowledgment of inter- and intra-tumoral heterogeneity and the dynamic and complex interactions within the tumor microenvironment (TME) [4,5,6,7], the necessity of simultaneously targeting a variety of different and complex signaling pathways has becomes evident. Consequently, complex combination or multimodal therapies are required [8]. Such approaches could enhance outcomes through their synergistic mechanisms of action by targeting the different properties of cancer cells or the TME. This creates a demand for alternative approaches in drug development [9, 10].

The amount of money invested in research and development (R&D) required to introduce a novel FDA-approved drug is continuously and dramatically increasing, while the likelihood for cancer drug approval in phase I clinical trials remains the lowest of any drug type at 6.7% [10,11,12]. This limitation in R&D productivity is termed as ‘Eroom’s Law’ [11]. Scannell et al. proposed four problems: (1) ‘the Beatles' problem’. In other words, developing new drugs that were significantly more efficient, safer, and improved over existing (and usually cheaper) therapies was like finding a music group better than the Beatles [13]; (2) the 'cautious regulator' problem is the increasing safety and formal requirements by the regulatory agencies [14,15,16,17]; (3) the 'throw money at it' problem occurs when the investment strategies try to overrate a potential new drug in order to improve the company competitiveness [18]; (4) and finally the 'basic research–brute force' bias that improvement of basic research and screening technology will always translate into effective drug discovery [18].

These limitations cannot be easily solved or controlled. Other challenges, however, are being addressed through the development of in vitro pharmacological profiling of drug candidates [19] and the integration of increasing basic research information into drug development pipeline design. This is particularly true in the human genome-wide association studies (GWASs) [20]. Furthermore, the careful consideration of the risk–benefit balance by regulatory agencies is also clearly important [21].

One strategy drug development limitations is the repurposing of already existing, clinically approved drugs for combinatory anticancer treatments [22,23,24]. This approach allows for the expedited development of novel therapies at a fraction of the costs and risks associated with novel drug discovery given that the safety profiles of these molecules have already been established. Furthermore, drug repurposing strategies are supported by progress in understanding both the mechanisms of human pathologies and the long-term consequences of these “old” drugs’ applications and by the continuous advancements in targeted drug delivery. Notably, the majority of currently marketed drugs have the ability to interact with more than one target, and occasionally include those that could benefit cancer patients [25,26,27].

Indeed, opportunistic findings led to the very first chemotherapeutics, chlorambucil (Leukeran) and busulfan (Myleran), which are still used to treat chronic lymphocytic and myeloid leukemias (CLL and CML) [28,29,30]. These drugs originated from ‘mustard gas’ and could be regarded as repurposed chemical weapons [31,32,33]. Nowadays, repurposing strategies often aim to substitute cytotoxic therapeutics with cytostatic ones, such as metformin and thalidomide, originally used to treat diabetes and inflammation, respectively [33]. Drug repurposing is not only limited to finding new uses for drugs initially developed for other diseases (including generics, on-patent drugs, and failed molecules), but also includes the original cancer drugs reassigned for different types of cancer or in different combinations.

Repurposing drugs can also be faster and less costly. For example, despite the need to test the new indications of repurposed drugs in later-phase clinical trials and obtaining approvals, knowing their safety profiles and pharmacokinetics/pharmacodynamics reduces the risk of failure [34, 35]. Although, the vast majority of molecules that undergo clinical trials do not make it to the clinic [36], some of those that fail at late stages could still be good candidates for repurposing and turn financial losses into potential successes.

In summary, drug repurposing strategies could improve the outcomes of modern mono and combinatory antitumor therapies. However, transitioning repurposed drugs into clinical application is accompanied by numerous commercial, technological, and regulatory obstacles. This short narrative review summarizes and discusses both the advantages and challenges associated with the repurposed drug component of antitumor therapeutic strategies.

Strategies for drug repurposing

Drug repurposing and is also known as drug repositioning or drug reprofiling. This classification can be further expanded to include failed drugs—compounds that have entered clinical trials but did not succeed due to unsatisfactory efficacy against the initial indication [37, 38]. All these compounds can be further divided into patent-protected (both approved and failed) and off-patent (generic) drugs. Obviously, the majority of research information comes from the latter group [37, 39, 40].

Although many successful drug reassignments have had rather serendipitous backgrounds, these favorable outcomes drive the optimistic and further development of systematic, dedicated strategies [41]. The simplest classification of drug discovery, including repurposing aspects, is divided into two basic lines of action: target-based drug discovery and phenotypic drug discovery [29]. The first approach starts by defining the molecular target underlying the pathology and aims to design dedicated drugs. For example, tamoxifen, which was initially aimed to be a contraceptive, was repurposed for the treatment of breast cancer since it was found to efficiently inhibit the estrogen receptor [42]. In contrast, phenotypic drug discovery ignores a specific drug target or hypothesis about its role in disease and instead tests candidates for desirable biological activity in ‘physiologically relevant’ systems. For instance, during a phenotypic screen for cell proliferation, it was found that auranofin, originally an anti-arthritic medication, effectively and selectively targets gastrointestinal stromal tumors, including imatinib-resistant ones [43, 44].

Drug repurposing strategies can also be classified into three main groups: target-centric, drug-centric, and disease-centric approaches [23]. Each of these strategies has its success stories, limitations, and advantages, and consequently, a dedicated group of supporters. The drug-centric approach focuses on identifying new indications for existing drugs, which can include expanding the existing license or patent towards novel off-label use of the compound in new medical conditions or groups of patients. This strategy is often applied to investigational or failed drugs that faced Eroom’s Law-related limitations in their initial assignment pipelines [23]. For example, valproic acid, originally indicated for bipolar disorder, has an off-target interaction with histone deacetylase 2, a protein that plays a role in many types of cancers. This has led to testing the repositioning of this drug for the treatment of neoplastic conditions such as familial adenomatous polyposis [45].

Drug-centric repositioning can be considered the least direct approach because the drug is only linked to a novel indication via the discovery of a target that is already established for this indication. Thus, a precise characterization of drug-target interactions is required to propose a novel repositioning hypothesis. The most common technical approaches for drug-centric repositioning are structure-based computational methods like molecular docking [46], pharmacophore modeling algorithms [47], protein–ligand interaction profile similarity testing [48], and machine learning approaches [49, 50]. However, since the drug needs to be repurposed to a novel target or disease, a structural model describing the binding mode of the drug to its original targets needs to be well defined. Furthermore, machine learning approaches are limited by the completeness of information in the databases that are utilized [20, 23, 51].

The target-centric approach matches a new indication without a treatment with an established drug and its known target. The old and new indications typically differ quite significantly. Complementary to a disease-centric approach, target-centric repositioning builds on a novel link between a new indication and an established target. It involves investigating the specific molecular targets implicated in the pathology of a disease and uses an existing drug proven to modulate those targets. For example, azacitidine, a potent inhibitor of DNA methyltransferases, was originally dedicated to treating myelodysplastic syndrome [52] and was later adapted for treating patients with acute myeloid leukemia and chronic myelomonocytic leukemia [52]. This approach is particularly useful when seeking to repurpose drugs to treat rare diseases.

Finally, the most effective approach so far is the disease-centric approach, which involves re-profiling drugs among different types of a disease, such as two types of cancer [23]. It involves identifying diseases with homologous underlying biological mechanisms and similar guiding principles to the indicated original drug treatments. For example, a drug developed to treat psoriasis could also treat other diseases with uncontrolled cell growth, such as cancer. In the case of cancer, these guiding principles are summarized in the Hallmarks of Cancer [53, 54]. Since these key hallmarks of malignancy are not regulated by a single signaling pathway [55], the pathways responsible for a cancer phenotype underlie the pathomechanisms of many other non-oncological human diseases. This opens the possibility of repurposing drugs towards novel anticancer therapeutics and supports agents in combinational approaches [56]. Notably, the diversity and complexity of the hallmarks of oncogenesis provide a strong rationale for using multiple drugs to obtain satisfactory therapy outcomes [40], while drug repositioning could significantly reduce costs and increase the availability of such novel therapies [56, 57].

Cancer complexity as a target of repurposed drugs

With the progress in understanding the molecular mechanisms related to oncogenesis, cancer progression, and treatments, more distinct attributes and signaling pathways are now recognized as crucial for various neoplastic diseases. Indeed, the initially proposed eight Hallmarks of Cancer have now been extended to fourteen [53, 54]. Although this diversity and complexity of neoplastic disease remain a therapeutic challenge, it also provides a strong rationale for drug repurposing. It is important to note that these hallmarks often result from the crosstalk between different signaling pathways, some of which are deregulated in cancer cells, while others compensate for this [58,59,60,61,62]. For example, cancer cells often efficiently avoid unfolded protein responses, hypoxia, and/or oxidative stress-related cell death signals and favor the proadaptive ones [63,64,65,66,67].

Nevertheless, the complexity of such signaling often limits the ability to selectively target cancer-specific signals without interfering those that are therapeutically desired. For example, inositol-requiring enzyme 1 (IRE1) activity, which is of great interest as a target for glioblastoma and triple-negative breast cancer, can lead to the accumulation of proadaptive signals. Whereas, at the same time, this enzyme can also support cell death signaling [7, 68,69,70]. Thus, despite the availability of both IRE1 inhibitors and potentiators, their translation into the clinic remains a challenge. Furthermore, healthy cells are often exposed to stress-inducing factors, with chemotherapeutics being one of them [71], and thus such stress-oriented therapies also carry the risks of adverse effects.

Chronic proliferation, a fundamental feature of cancer cells, often results from a network of deregulated signaling pathways and growth factors. These pathways operate mainly by receiving signals from growth factors that bind to cell-surface receptors typically with intracellular tyrosine kinase activity [72]. This activity modulates pro-survival signaling pathways such as phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), the mammalian target of rapamycin (mTOR) [73] and the mitogen-activated protein kinases/extracellular signal-regulated kinase (MAPK/ERK) [74]. The development of specific receptor tyrosine kinase inhibitors has resulted in effective chemotherapeutics [74]. However, cancer cells can also utilize many other pathways to proliferate and eventually circumvent the inhibited routes in order to develop resistance [75, 76]. Repurposed drugs may provide a solution to this problem. For example, rapamycin, an mTOR1 inhibitor initially approved as an immunosuppressant and later as an anti-restenosis agent [77, 78], was repurposed to treat leukemias due to the importance of the mTOR pathway in cancer [79, 80] [81]. However, inhibition of mTOR1 is often compensated by the activation of PI3K-AKT [82] and the reactivation of eukaryotic translation initiation factor 4E-binding proteins [83, 84]. Despite these limitations, rapamycin and its more soluble and specific analogs, like temsirolimus [85], have been tested in combination with growth factor receptor antagonists [86].

Other candidates for repurposing that could target cancer proliferation include prazosin, an alpha blocker initially approved to treat hypertension [87], and indomethacin, a non-steroidal anti-inflammatory drug (NSAID) [88]. Prazosin has been reported to inhibit AKT signaling [89, 90] and is recommended for treating pheochromocytoma [91]. It is also included in a Phase 1 study as an additive to radiotherapy in men with prostate cancer (ACTRN12621000784819). Indomethacin, besides its Cox1/2 inhibition-related antiangiogenic effects [92], has been shown to impair cancer proliferation by inhibiting MAPK [93] or PKC signaling [94]. Currently, three indomethacin-related clinical trials are registered, including a Phase 4 trial for prostate cancer (ChiCTR2000038968) and a Phase 1 study for breast cancer (NCT02950259). Furthermore, drugs that could effectively target human telomerase reverse transcriptase (hTERT) in cancer cells to limit their replication remain an interest for drug repurposing strategies [95].

Another promising strategy is the search for drugs that can accelerate cancer cell death. Along with progress in understanding the molecular mechanisms underlying regulated cell death [96, 97], many related adverse effects of non-oncological drugs may be useful in cancer therapies. Considering that cancer cells often circumvent common apoptotic pathways, drugs that can selectively accelerate other cell death modalities—including ferroptosis [98], cuproptosis [99], necroptosis [100], pyroptosis[101], and lethal autophagy [102]—can improve treatment strategies while reducing general toxicity. One exception, however, is autophagy since this can also favor cancer cells by enhancing their survival under metabolic and environmental stresses [103], and therefore should be considered with caution.

Artemisinin and chloroquine, along with their derivatives were initially dedicated for treating malaria [104], are well-known examples of cell death-related drug repurposing. Artemisinins have been reported to induce non-apoptotic programmed cell death, especially ferroptosis, in cancer cells [105,106,107,108,109]. Recent clinical trials for artesunate are testing its application in colorectal cancers (NCT02633098) and leukemias (CTRI/2024/03/063617). Chloroquine and its derivative hydroxychloroquine are approved as autophagy flux inhibitors to treat pancreatic and other cancers [110, 111]. Currently, these compounds are undergoing 48 clinical trials related to cancer therapies (Database Repurposing Trials In Oncology, ReDO_Trials_DB, https://www.anticancerfund.org/en/database-repurposing-trials-oncology as for 14.08.2024) [112].

Mebendazole (5-benzoyl-1H-benzimidazol-2-ylcarbamate), initially intended as an anthelmintic agent [113], is another candidate for repurposing into oncological therapies due to its potential to inhibit microtubule polymerization [114, 115]. Notably, besides restricting tumor growth, this compound has been effective in preventing the invasion and metastasis of malignant tumors and glioblastomas [116, 117]. Zhang [118] and in other individual cancer cases [114, 115]. Currently, six mebendazole-related clinical trials are registered in the ReDO_Trials_DB [112].

Deregulated metabolism and increased energy demands are other hallmarks of cancer [119, 120] that have been targeted for drug repurposing [121]. Patients with diabetes are generally more prone to several types of cancer [122, 123]. Long-term treatment with metformin, approved for obese type 2 diabetes [124], has been observed to lower the risk of cancer in diabetic patients, making this drug a potential candidate for repurposing [120, 122]. Indeed, there are 133 metformin-related trials in the ReDO_Trials_DB [112]. However, despite these efforts, results have been far from satisfactory [125, 126]. Furthermore, the mechanism of action of metformin in tumor cells and the tumor microenvironment remains unclear and under discussion. Although high doses of this compound impair cellular respiration by inhibiting Complex-1 [127], preclinical studies have observed a plethora of pleiotropic effects of metformin administration in cancer cells that are independent of Complex-1 inhibition [128, 129]. Hopefully, continuous research on this compound will eventually allow successful metformin repurposing.

Disulfiram provides another example of a repurposed drug, initially intended to treat alcoholism [130], now targeting cancer cell metabolism 129,130. Disulfiram inhibits acetaldehyde dehydrogenase activity [131], resulting in alcohol intolerance, as well as the blockage of formaldehyde oxidation [132] and deregulated oxidative metabolism in cancer cells [133]. Furthermore, the p97 segregase adaptor NPL4 (also known as VCP), important for maintaining cellular proteostasis, has been identified as the molecular target of disulfiram responsible for its anticancer activity [134]. Currently, there are 13 disulfiram clinical trials in the ReDO_Trials_DB, including a phase 2/3 application of this drug for glioblastoma (NCT02678975) [112].

The ability of cancers to impair and circumvent host immune responses provides another therapeutic opportunity [135,136,137,138]. Pharmaceutical solutions that could increase antitumor immunity, such as immune checkpoint inhibitors, are currently of great interest [139]. It has been observed that some vaccines dedicated to infectious diseases (such as rotaviruses, yellow fever, and influenza), when administered intratumorally, can activate antitumor immunity [140,141,142,143].

Other drug repurposing strategies arise from the specific impairment of tumor suppressors such as p53 or the retinoblastoma protein in cancer cells [144]. Along these lines, quinacrine, an antimalarial agent [145], was found to be a promising candidate since it was reported to induce p53 expression in cancer cells [146] and can exert some anticancer activity in a p53-dependent manner [146, 147]. The accumulation of p53 has also been observed in cancer cells treated with ritonavir, a protease inhibitor used to treat human immunodeficiency virus (HIV) infection [148, 149]. Furthermore, other reports found this compound capable of reactivating the retinoblastoma protein [147]. Currently, there is one active phase 1 clinical trial for the application of ritonavir for prostate cancer (NCT05679388) [112]. Notably, statins have also been shown to increase p53 activity in cancer cells and thus display anticancer potential [148, 149]. Currently, there are 47 statin-related records in the ReDO_Trials_DB, many of which are reaching phase 3 or 4 [112].

Tumor expansion is accompanied by an increased demand for nutrients and oxygen by cancer cells, leading to the induction of chronic angiogenesis [150]. Although this hallmark of cancer cells has resulted in the development of currently used antiangiogenic agents that limit tumor blood flow and lead to its starvation [58, 151], the use of these drugs is limited and can unfortunately stimulate resistance [152]. Notably, the repurposing of thalidomide, an immunomodulatory drug initially sold to treat morning sickness that was withdrawn worldwide in 1962 after it was linked to severe birth defects [153], has shown promise. Thalidomide still remains in use to treat leprosy [153] and is currently used as an antiangiogenic agent [154, 155]. It is also approved for combination therapy in multiple myeloma [156].

Antiangiogenic and anticancer potential has also been observed for the approved antifungal agent itraconazole [157, 158]. Currently, there are 17 itraconazole-related records in the ReDO_Trials_DB, one of which for ovarian cancer is reaching phase 3 (NCT03458221). Interestingly, artemisinins have also been assigned antiangiogenic activities [159]. In contrast, it has been reported that some anticancer approaches aim to induce angiogenesis in order to facilitate drugs delivery [160, 161].

Notably, cancer-related inflammation supports not only angiogenesis but also invasion and metastasis, as well as reprogramming of the tumor microenvironment (TME) [162, 163]. Therefore, anti-inflammatory drugs could be good candidates for repurposing. Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor approved for adult arthritis [164], is extensively being tested in clinical trials (57 registered in ReDO_Trials_DB) due to multiple reports of its potential to enhance the chemosensitivity of cancer cells and to reduce the toxicity of marketed chemotherapeutics [165,166,167]. Similarly, 31 clinical trials are registered for aspirin (ReDO_Trials_DB), which was already suggested to have potential as an anticancer drug in the 1970s [168]. More recent research reports of aspirin being effective against many types of cancer [169,170,171] are supported by epidemiological analyses [172,173,174,175]. Indeed, low-dose aspirin inhibits the production of thromboxane A2 (TXA2) by irreversibly inhibiting the enzyme COX-1 in platelets. By reducing TXA2, aspirin can help prevent the formation of blood clots and has been shown to have potential benefits in reducing the risk of cancer progression and metastasis [176, 177]. Furthermore, aspirin at higher doses is also a more potent COX-2 inhibitor, which can increase its anti-cancer properties against tumors that overexpress this enzyme [175]. Numerous studies shown that daily low-doses of aspirin may significantly reduce the risk of colon cancer and rectal cancer [178] and breast cancer [179, 180]. Recent results from a 20-year cohort study involving 1,909,531 individuals in Denmark have shown that long-term low-dose aspirin use is associated with slightly to moderately reduced risks for several specific cancers [181]. However, there was no reduction in overall cancer risk for some common cancers [181]. Similar or slightly stronger inverse associations were observed for the consistent use of high-dose aspirin [181].

Interestingly, the ASPirin in Reducing Events in the Elderly (ASPREE) study, which was double-blind and performed on a large cohort (for 4.7 years and over 19,000 individuals older than 65–70, that did not have cardiovascular disease, dementia, or disability), showed no advantage of taking low-dose aspirin, and in fact increased the risk of being diagnosed with stage 3 or 4 cancers as well as increased mortality rates compared to the placebo [182]. In contrast, prior randomized controlled trials, mainly involving younger individuals, demonstrated a delayed cancer benefit with aspirin [182]. However, these study conclusions are under discussion and should be taken with caution, as deaths were classified according to the underlying cause by adjudicators who were unaware of the trial-group assignments. Furthermore, hazard ratios were calculated to compare mortality between the aspirin group and the placebo group, and post hoc exploratory analyses of specific causes of death were performed [182].

Furthermore, some of the adverse effects of many approved chemotherapeutics can be reduced with the use of additional anti-inflammatory compounds and beta-blockers to reduce cardiotoxicity [183, 184]. The latter (especially propranolol and timolol) have been reported to have anticancer activity [185,186,187]. Taken together, these examples provided illustrate the vast potential of drug repurposing in oncology.

Discussion

Although the drug repurposing approach seems like an attractive solution to benefit both cancer patients and health systems, its results have been below expectations despite the considerable efforts and high academic attention. Continuous research and technological development have yet to yield breakthrough solutions. While there are some examples of successful drug repositioning, the majority of candidates, like metformin, have remained in the reassignment pipelines for many years. As of August 14, 2024, the ReDO_Trials_DB database reports 898 (409 controlled) trials related to 182 drugs (with metformin being the most commonly tested one) and involving 157,295 patients [112] (Fig. 1). Notably, however, less than 5% of these trials are sponsored by pharmaceutical companies, with the vast majority funded by non-profit organizations [188, 189]. Furthermore, 139 clinical trials have reached phase 3 or 4 [112].

Fig. 1
figure 1

A. Repurposing trials are a modest percentage of all oncology clinical trials. B The most popular drugs in repurposing trials (C) and the most popular cancers to be targeted in controlled trails of repurposed drugs. The data were obtained from the ReDO_Trials_DB database and the GlobalData’s Clinical Trials Intelligence Center (December 15, 2023)

Although these numbers may initially seem impressive, they are modest compared to data from the GlobalData’s Clinical Trials Intelligence Center ((https://www.clinicaltrialsarena.com/sponsored/oncology-in-2024-the-clinical-trial-trends-reshaping-the-role-of-cros/). As of December 15, 2023, 6,528 oncology-related trials were noted, with another 569 planned to begin. In the DrugBank database ((https://go.drugbank.com/releases/latest), there are 4,493 approved drugs (2812 small molecules and 1681 biotechnology drugs) that can be directed against over 3,000 unique targets (Fig. 1). Additionally, there are over 333 withdrawn drugs (302 small molecules and 31 biotechnology drugs), 8,000 investigational ones (5311 small molecules and 2752 biotechnology drugs), and about 6732 experimental compounds (6353 small molecules and 379 biotechnology drugs). Taken together, drug repurposing in oncology, despite its potential and large number of possible candidates, remains underappreciated and mostly limited to academic research and small biotech companies.

Notably, the business models of Big Pharma rely strongly on market exclusivity for their drugs, allowing them to sell their drugs at high prices [190,191,192]. Since repurposed drugs cannot usually be considered novel chemical entities and their structures are already known, novel patent claims to the active pharmaceutical ingredient are not possible [193]. The repurposed drug can only be protected at the level of the ‘method of use’ for the new indication, although such protection is harder to obtain and costs more [193]. Finally, the use of patents excludes off-label prescriptions, where medications are prescribed for indications or populations for which they have no regulatory approval [194]. Numerous solutions have been suggested to motivate Big Pharma efforts towards repurposing (tax breaks, FDA-priority review vouchers, or funding clinical trials) [195, 196]. However, given the current business model Big Pharma operates under, implementing these initiatives probably will not be a game changer [188]. This conflict of interest is well illustrated by thalidomide approvals. Despite this drug in combination with melphalan–prednisone is comparable to the dramatically more expensive lenalidomide, the more expensive new drug was approved as the standard therapy [197, 198].

Indeed, mainly academic and independent research provides the rationale for using off-patent medications in cancer treatment [40]. However, in the case of drug repurposing, these academic approaches often lack specific insights that are exclusive to pharmaceutical companies. Due to limited resources, lack of data, technology, funding, and experience, many academic attempts at drug repurposing are often “fashion” driven (metformin, statins, aspirin, ascorbic acid, etc.) [112] and thus oriented on drugs that may be easy to obtain, publish, and get funded for their application (Fig. 1B). However, this approach does not consider that millions of people who are cancer patients or will develop cancers are already taking these prescriptions, whereas their benefits on a wide population scale in terms of cancer risks are usually under discussion or not properly documented.

Furthermore, academic research is the main contributor of related omics and epidemiological data that are further used for machine learning and other applications utilized by drug repositioning strategies. Notably, retrospective observational studies are subject to immortal time bias and selection bias [199], resulting in frequent overestimations of their advantages for the treatment group [200]. This case is well illustrated by the numerous correlations between metformin treatments and the incidence of cancer [201]. Furthermore, since metformin’s original target group were diabetic patients who differ from cancer patients, selection bias occurred [202]. Additionally, computational strategies for predicting drug reassignment are only as good as the reliability of the input data and can often discourage target-centric and drug-centric strategies. Phenotype-based high throughput screening strategies, due to the costs of libraries and extensive labor, are usually beyond basic research funding schemes. Finally, the costs and ability to design dedicated and sufficient clinical trials that can include various cancer types and patient groups, remain another serious barrier for academic approaches.

The complexity of cancer limits the efficacy of monotherapies, whereas combination therapies come with specific challenges and limitations that often translate into drug repurposing approaches [199, 203, 204]. Notably, combination therapy trials are more complex and thus cost more than those for monotherapies [205]. Since phase 2 trials are usually the turning point for a drug’s fate, progressing them towards more randomized larger cohort research trials requires promising efficacy results in order to justify the financial burden [199].

Many clinical trials fail due to insufficient cancer patient accrual [206]. Cancer patients are often older people with accompanying diseases and undergoing different cancer-related or unrelated treatments, which may result in unexpected side effects and death during the trials [33, 207]. Indeed, combinational therapies do not guarantee better efficacy [33]. For example, combined dacarbazine with cisplatin, carmustine, and tamoxifen for metastatic (stage IV) melanoma treatment is comparable in terms of patient survival with monotherapy based on high-dose dacarbazine [207, 208]. Unfortunately, dosages that may be well tolerated in trials conducted using healthy subjects or when used for treating the diseases the drug was originally intended for may not be achievable in cancer patients [33]. Repurposed drugs intended for specific targets might not have the same efficacy in cancer cells or in the presence of other drugs [33]. Therefore, their repositioning may require higher doses, which can result in novel distinct mechanisms of action and consequently unforeseen adverse effects [209, 210]. For example, aspirin repurposed for use in high doses may lead to an increased risk of gastrointestinal bleeding, while simvastatin and metformin contribute to the development of hypolipidemia and hypoglycemia. All these issues stress the importance of quality preclinical research that will allow only the most effective and safest combinations to be trialed in phases 2 and 3 [204, 211, 212].

Importantly, recent scientific and technological breakthroughs are becoming more economically available and thus possible to incorporate into high-throughput screening pipelines, as well as drug-centric approaches. Starting with omics approaches, which allow the determination of molecular mechanisms of repurposing candidates, and coupling this with failed drugs could provide valuable insight into the cellular proteomic, metabolomic, and transcriptomic changes [44, 213,214,215,216,217,218,219,220,221,222] in various cancers. Importantly, single-cell sequencing seems to be a way to address cancer heterogeneity [223], while the rapid growth and development of databases provide machine learning and computational approaches with more reliable insights [224,225,226,227]. Furthermore, the development of novel drug delivery methods that allow more specific and even compartment-targeted application of repurposed drugs may contribute to the success of novel repurposing strategies [228,229,230].

Conclusions and perspectives

Repurposed drugs have the undeniable potential to address the shortage of new drugs and combat acquired chemotherapy resistance. Additionally, many healthcare systems struggle to provide patients with expensive new generations of chemotherapeutics, let alone personalized therapies. The financial advantages of drug repositioning could benefit many patients worldwide. However, despite research progress, multiple pharmacological challenges and obstacles need to be addressed to effectively utilize the opportunity of repurposed drugs in cancer treatment. Importantly, the field requires programs, regulations, and government-level funding to promote and support collaboration between academia and the pharmaceutical industry [40, 190]. Such programs in the USA and UK have allowed the transfer of a large number of failed drugs to academic research for repurposing [231]. Furthermore, continuous efforts and lobbying by academic and non-profit organizations are necessary to popularize and develop new drug discovery concepts.

Availability of data and materials

Not applicable.

Abbreviations

AKT:

Protein kinase B

ASPREE:

ASPirin in Reducing Events in the Elderly

CLL:

Chronic lymphocytic leukemia

CML:

Chronic myeloid leukemia

COX-2:

Cyclooxygenase-2

ERK:

Extracellular signal-regulated kinase

FDA:

Food and Drug Administration

GWASs:

Genome-wide association studies

HIV:

Human immunodeficiency virus

hTERT:

Human telomerase reverse transcriptase

MAPK:

Mitogen-activated protein kinases

mTOR:

Mammalian target of rapamycin

NSAID:

Non-steroidal anti-inflammatory drug

PI3K:

Phosphatidylinositol 3-kinase

R&D:

Research and development

TME:

Tumor microenvironment

References

  1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clin. 2024;74(3):229–63.

    Google Scholar 

  2. Global Burden of Disease Cancer C. The global burden of cancer 2013. JAMA Oncol. 2015;1(4):505–27.

    Article  Google Scholar 

  3. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–26.

    Article  PubMed  CAS  Google Scholar 

  4. Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. 2020;30(16):R921–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Communi Signal. 2020;18(1):59.

    Article  Google Scholar 

  6. de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41(3):374–403.

    Article  PubMed  Google Scholar 

  7. Bartoszewska S, Collawn JF, Bartoszewski R. The role of the hypoxia-related unfolded protein response (UPR) in the tumor microenvironment. Cancers. 2022;14(19):4870.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Qin S-Y, Cheng Y-J, Lei Q, Zhang A-Q, Zhang X-Z. Combinational strategy for high-performance cancer chemotherapy. Biomaterials. 2018;171:178–97.

    Article  PubMed  CAS  Google Scholar 

  9. Vokinger KN, Hwang TJ, Grischott T, Reichert S, Tibau A, Rosemann T, Kesselheim AS. Prices and clinical benefit of cancer drugs in the USA and Europe: a cost-benefit analysis. Lancet Oncol. 2020;21(5):664–70.

    Article  PubMed  CAS  Google Scholar 

  10. Sun D, Gao W, Hu H, Zhou S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm Sin B. 2022;12(7):3049–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Scannell JW, Blanckley A, Boldon H, Warrington B. Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov. 2012;11(3):191–200.

    Article  PubMed  CAS  Google Scholar 

  12. Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32(1):40–51.

    Article  PubMed  CAS  Google Scholar 

  13. Milsted RA. Cancer drug approval in the United States, Europe, and Japan. Adv Cancer Res. 2007;96:371–91.

    Article  PubMed  CAS  Google Scholar 

  14. Bateman-House A, Robertson CT. The federal right to try act of 2017-A wrong turn for access to investigational drugs and the path forward. JAMA Intern Med. 2018;178(3):321–2.

    Article  PubMed  Google Scholar 

  15. Khozin S, Liu K, Jarow JP, Pazdur R. Regulatory watch: why do oncology drugs fail to gain US regulatory approval? Nat Rev Drug Discov. 2015;14(7):450–1.

    Article  PubMed  CAS  Google Scholar 

  16. Benderly BL. Experimental drugs on trial. Sci Am. 2007;297(4):92–9.

    Article  PubMed  Google Scholar 

  17. FDA treads delicate line between safety and speed. Oncology (Williston Park). 1999;13(1):16

  18. Iazzolino G, Bozzo R. Partnership models for R & D in the pharmaceutical industry. In: Canci JK, Mekler P, Mu G, editors. Quantitative models in life science business: from value creation to business processes. Cham: Springer International Publishing; 2023. p. 29–48.

    Chapter  Google Scholar 

  19. Bowes J, Brown AJ, Hamon J, Jarolimek W, Sridhar A, Waldron G, Whitebread S. Reducing safety-related drug attrition: the use of in vitro pharmacological profiling. Nat Rev Drug Discov. 2012;11(12):909–22.

    Article  PubMed  CAS  Google Scholar 

  20. Kang H, Pan S, Lin S, Wang Y-Y, Yuan N, Jia P. PharmGWAS: a GWAS-based knowledgebase for drug repurposing. Nucleic Acids Res. 2024;52(D1):D972–9.

    Article  PubMed  CAS  Google Scholar 

  21. Ringel MS, Scannell JW, Baedeker M, Schulze U. Breaking Eroom’s Law. Nat Rev Drug Discov. 2020;19(12):833–4.

    Article  PubMed  CAS  Google Scholar 

  22. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.

    Article  PubMed  CAS  Google Scholar 

  23. Parisi D, Adasme MF, Sveshnikova A, Bolz SN, Moreau Y, Schroeder M. Drug repositioning or target repositioning: a structural perspective of drug-target-indication relationship for available repurposed drugs. Comput Struct Biotechnol J. 2020;18:1043–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, et al. Repurposing of drugs: an attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol. 2021;68:258–78.

    Article  PubMed  CAS  Google Scholar 

  25. Huang A, Garraway LA, Ashworth A, Weber B. Synthetic lethality as an engine for cancer drug target discovery. Nat Rev Drug Discov. 2020;19(1):23–38.

    Article  PubMed  CAS  Google Scholar 

  26. Dallavalle S, Dobričić V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, et al. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat. 2020;50:100682.

    Article  PubMed  Google Scholar 

  27. Patel MN, Halling-Brown MD, Tym JE, Workman P, Al-Lazikani B. Objective assessment of cancer genes for drug discovery. Nat Rev Drug Discov. 2013;12(1):35–50.

    Article  PubMed  CAS  Google Scholar 

  28. Eder J, Sedrani R, Wiesmann C. The discovery of first-in-class drugs: origins and evolution. Nat Rev Drug Discov. 2014;13(8):577–87.

    Article  PubMed  CAS  Google Scholar 

  29. Swinney DC. Phenotypic vs. target-based drug discovery for first-in-class medicines. Clin Pharmacol Ther. 2013;93(4):299–301.

    Article  PubMed  CAS  Google Scholar 

  30. Moffat JG, Vincent F, Lee JA, Eder J, Prunotto M. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat Rev Drug Discov. 2017;16(8):531–43.

    Article  PubMed  CAS  Google Scholar 

  31. Adair FE, Bagg HJ. Experimental and clinical studies on the treatment of cancer by dichlorethylsulphide (mustard gas. Ann Surg. 1931;93(1):190–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Haddow A. On the biological alkylating agents. Perspect Biol Med. 1973;16(4):503–24.

    Article  PubMed  CAS  Google Scholar 

  33. Schein CH. Repurposing approved drugs for cancer therapy. Br Med Bull. 2021;137(1):13–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Maxmen A. Busting the billion-dollar myth: how to slash the cost of drug development. Nature. 2016;536(7617):388–90.

    Article  PubMed  CAS  Google Scholar 

  35. Bertolini F, Sukhatme VP, Bouche G. Drug repurposing in oncology–patient and health systems opportunities. Nat Rev Clin Oncol. 2015;12(12):732–42.

    Article  PubMed  Google Scholar 

  36. Mullard A. Biotech R&D spend jumps by more than 15. Nat Rev Drug Discov. 2016;15(7):447.

    PubMed  Google Scholar 

  37. Weir SJ, DeGennaro LJ, Austin CP. Repurposing approved and abandoned drugs for the treatment and prevention of cancer through public-private partnership. Cancer Res. 2012;72(5):1055–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Fogel DB. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contemp Clin Trials Commun. 2018;11:156–64.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Cha Y, Erez T, Reynolds IJ, Kumar D, Ross J, Koytiger G, et al. Drug repurposing from the perspective of pharmaceutical companies. Br J Pharmacol. 2018;175(2):168–80.

    Article  PubMed  CAS  Google Scholar 

  40. Weth FR, Hoggarth GB, Weth AF, Paterson E, White MPJ, Tan ST, et al. Unlocking hidden potential: advancements, approaches, and obstacles in repurposing drugs for cancer therapy. Br J Cancer. 2024;130(5):703–15.

    Article  PubMed  Google Scholar 

  41. Moffat JG, Vincent F, Lee JA, Eder J, Prunotto M. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat Rev Drug Discov. 2017;16(8):531–43.

    Article  PubMed  CAS  Google Scholar 

  42. Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205–13.

    Article  PubMed  CAS  Google Scholar 

  43. Pessetto ZY, Weir SJ, Sethi G, Broward MA, Godwin AK. Drug repurposing for gastrointestinal stromal tumor. Mol Cancer Ther. 2013;12(7):1299–309.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Macioszek S, Dudzik D, Bartoszewski R, Stokowy T, Lambrechts D, Boeckx B, et al. Metabolomic and transcriptomic response to imatinib treatment of gastrointestinal stromal tumour in xenograft-bearing mice. Transl Oncol. 2023;30:101632.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Huang X, Guo B. Adenomatous polyposis coli determines sensitivity to histone deacetylase inhibitor-induced apoptosis in colon cancer cells. Can Res. 2006;66(18):9245–51.

    Article  CAS  Google Scholar 

  46. Meng XY, Zhang HX, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des. 2011;7(2):146–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Giordano D, Biancaniello C, Argenio MA, Facchiano A. Drug design by pharmacophore and virtual screening approach. Pharmaceuticals. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph15050646.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Salentin S, Schreiber S, Haupt VJ, Adasme MF, Schroeder M. PLIP: fully automated protein–ligand interaction profiler. Nucleic Acids Res. 2015;43(W1):W443–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Yan X-Y, Zhang S-W, Zhang S-Y. Prediction of drug–target interaction by label propagation with mutual interaction information derived from heterogeneous network. Mol BioSyst. 2016;12(2):520–31.

    Article  PubMed  CAS  Google Scholar 

  50. Catacutan DB, Alexander J, Arnold A, Stokes JM. Machine learning in preclinical drug discovery. Nat Chem Biol. 2024;20(8):960–73.

    Article  PubMed  CAS  Google Scholar 

  51. Pahikkala T, Airola A, Pietilä S, Shakyawar S, Szwajda A, Tang J, Aittokallio T. Toward more realistic drug-target interaction predictions. Brief Bioinform. 2015;16(2):325–37.

    Article  PubMed  CAS  Google Scholar 

  52. Gore SD. New ways to use DNA methyltransferase inhibitors for the treatment of myelodysplastic syndrome. Hematol Am Soc Hematol Educ Program. 2011;2011:550–5.

    Article  Google Scholar 

  53. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  PubMed  CAS  Google Scholar 

  54. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46.

    Article  PubMed  CAS  Google Scholar 

  55. Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the hallmarks of cancer. Science. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.aal2380.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Sun W, Sanderson PE, Zheng W. Drug combination therapy increases successful drug repositioning. Drug Discov Today. 2016;21(7):1189–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Boshuizen J, Peeper DS. Rational cancer treatment combinations: an urgent clinical need. Mol Cell. 2020;78(6):1002–18.

    Article  PubMed  CAS  Google Scholar 

  58. Slawski J, Jaśkiewicz M, Barton A, Kozioł S, Collawn JF, Bartoszewski R. Regulation of the HIF switch in human endothelial and cancer cells. Eur J Cell Biol. 2024;103(2):151386.

    Article  PubMed  CAS  Google Scholar 

  59. Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim et Biophys Acta (BBA) – Mol Cell Biol Lip. 2024;1869(7):159515.

    Google Scholar 

  60. Bartoszewska S, Cabaj A, Dąbrowski M, Collawn JF, Bartoszewski R. miR-34c-5p modulates X-box-binding protein 1 (XBP1) expression during the adaptive phase of the unfolded protein response. Faseb J. 2019;33(10):11541–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Bartoszewski R, Gebert M, Janaszak-Jasiecka A, Cabaj A, Króliczewski J, Bartoszewska S, et al. Genome-wide mRNA profiling identifies RCAN1 and GADD45A as regulators of the transitional switch from survival to apoptosis during ER stress. FEBS J. 2020;287(14):2923–47.

    Article  PubMed  CAS  Google Scholar 

  62. Gebert M, Sobolewska A, Bartoszewska S, Cabaj A, Crossman DK, Króliczewski J, et al. Genome-wide mRNA profiling identifies X-box-binding protein 1 (XBP1) as an IRE1 and PUMA repressor. Cell Mol Life Sci. 2021;78(21):7061–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Payne KK. Cellular stress responses and metabolic reprogramming in cancer progression and dormancy. Semin Cancer Biol. 2022;78:45–8.

    Article  PubMed  CAS  Google Scholar 

  64. Aranda-Anzaldo A, Dent MAR. Is cancer a disease set up by cellular stress responses? Cell Stress Chaperones. 2021;26(4):597–609.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Fulda S, Gorman AM, Hori O, Samali A. Cellular stress responses: cell survival and cell death. Int J Cell Biol. 2010;2010(1):214074.

    PubMed  PubMed Central  Google Scholar 

  66. Moszyńska A, Collawn JF, Bartoszewski R. IRE1 endoribonuclease activity modulates hypoxic HIF-1α signaling in human endothelial cells. Biomolecules. 2020;10(6):895.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Moszyńska A, Jaśkiewicz M, Serocki M, Cabaj A, Crossman DK, Bartoszewska S, et al. The hypoxia-induced changes in miRNA-mRNA in RNA-induced silencing complexes and HIF-2 induced miRNAs in human endothelial cells. Faseb J. 2022;36(7):e22412.

    Article  PubMed  Google Scholar 

  68. Bartoszewska S, Sławski J, Collawn JF, Bartoszewski R. Dual RNase activity of IRE1 as a target for anticancer therapies. J Cell Communi Signal. 2023;17(4):1145–61.

    Article  CAS  Google Scholar 

  69. Gebert M, Bartoszewska S, Opalinski L, Collawn JF, Bartoszewski R. IRE1-mediated degradation of pre-miR-301a promotes apoptosis through upregulation of GADD45A. Cell Commun Signal. 2023;21(1):322.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Gebert M, Sławski J, Kalinowski L, Collawn JF, Bartoszewski R. The unfolded protein response: a double-edged sword for brain health. Antioxidants. 2023;12(8):1648.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Tiligada E. Chemotherapy: induction of stress responses. Endocr Relat Cancer. 2006;13(Suppl 1):S115–24.

    Article  PubMed  CAS  Google Scholar 

  72. Du Z, Lovly CM. Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 2018;17(1):58.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Panwar V, Singh A, Bhatt M, Tonk RK, Azizov S, Raza AS, et al. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct Target Ther. 2023;8(1):375.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Pottier C, Fresnais M, Gilon M, Jérusalem G, Longuespée R, Sounni NE. Tyrosine kinase inhibitors in cancer: breakthrough and challenges of targeted therapy. Cancers. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers12030731.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, Poulikakos PI, Scaltriti M, Moskatel E, et al. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov. 2011;1(3):248–59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Ahronian LG, Sennott EM, Van Allen EM, Wagle N, Kwak EL, Faris JE, et al. Clinical acquired resistance to RAF inhibitor combinations in BRAF-mutant colorectal cancer through MAPK pathway alterations. Cancer Discov. 2015;5(4):358–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Benjamin D, Colombi M, Moroni C, Hall MN. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov. 2011;10(11):868–80.

    Article  PubMed  CAS  Google Scholar 

  78. Farb A, John M, Acampado E, Kolodgie FD, Prescott MF, Virmani R. Oral everolimus inhibits in-stent neointimal growth. Circulation. 2002;106(18):2379–84.

    Article  PubMed  CAS  Google Scholar 

  79. Grabiner BC, Nardi V, Birsoy K, Possemato R, Shen K, Sinha S, et al. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov. 2014;4(5):554–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Altman JK, Sassano A, Kaur S, Glaser H, Kroczynska B, Redig AJ, et al. Dual mTORC2/mTORC1 targeting results in potent suppressive effects on acute myeloid leukemia (AML) progenitors. Clin Cancer Res. 2011;17(13):4378–88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Sillaber C, Mayerhofer M, Böhm A, Vales A, Gruze A, Aichberger K, et al. Evaluation of antileukaemic effects of rapamycin in patients with imatinib-resistant chronic myeloid leukaemia. Eur J Clin Invest. 2008;38(1):43–52.

    Article  PubMed  CAS  Google Scholar 

  82. O’Reilly KE, Rojo F, She Q-B, Solit D, Mills GB, Smith D, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Can Res. 2006;66(3):1500–8.

    Article  CAS  Google Scholar 

  83. Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A. 2008;105(45):17414–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Dowling RJ, Topisirovic I, Alain T, Bidinosti M, Fonseca BD, Petroulakis E, et al. mTORC1-mediated cell proliferation, but not cell growth, controlled by the 4E-BPs. Science. 2010;328(5982):1172–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Malizzia LJ, Hsu A. Temsirolimus, an mTOR inhibitor for treatment of patients with advanced renal cell carcinoma. Clin J Oncol Nurs. 2008;12(4):639–46.

    Article  PubMed  Google Scholar 

  86. Rugo HS, Trédan O, Ro J, Morales SM, Campone M, Musolino A, et al. A randomized phase II trial of ridaforolimus, dalotuzumab, and exemestane compared with ridaforolimus and exemestane in patients with advanced breast cancer. Breast Cancer Res Treat. 2017;165:601–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Kirkendall WM, Hammond JJ, Thomas JC, Overturf ML, Zama A. Prazosin and clonidine for moderately severe hypertension. JAMA. 1978;240(23):2553–6.

    Article  PubMed  CAS  Google Scholar 

  88. Percy J, Stephenson P, Thompson M. Indomethacin in the treatment of rheumatic diseases. Ann Rheum Dis. 1964;23(3):226.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Desiniotis A, Kyprianou N. Advances in the design and synthesis of prazosin derivatives over the last ten years. Expert Opin Ther Targets. 2011;15(12):1405–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Assad Kahn S, Costa SL, Gholamin S, Nitta RT, Dubois LG, Fève M, et al. The anti-hypertensive drug prazosin inhibits glioblastoma growth via the PKC δ-dependent inhibition of the AKT pathway. EMBO Mol Med. 2016;8(5):511–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Nicholson JP Jr, Vaughn ED Jr, Pickering TG, Resnick LM, Artusio J, Kleinert HD, et al. Pheochromocytoma and prazosin. Ann Internal Med. 1983;99(4):477–9.

    Article  Google Scholar 

  92. Touhey S, O’Connor R, Plunkett S, Maguire A, Clynes M. Structure–activity relationship of indomethacin analogues for MRP-1, COX-1 and COX-2 inhibition: identification of novel chemotherapeutic drug resistance modulators. Eur J Cancer. 2002;38(12):1661–70.

    Article  PubMed  CAS  Google Scholar 

  93. Lin C-C, Suen KM, Stainthorp A, Wieteska L, Biggs GS, Leitão A, et al. Targeting the Shc-EGFR interaction with indomethacin inhibits MAP kinase pathway signalling. Cancer Lett. 2019;457:86–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Mazumder S, De R, Debsharma S, Bindu S, Maity P, Sarkar S, et al. Indomethacin impairs mitochondrial dynamics by activating the PKCζ–p38–DRP1 pathway and inducing apoptosis in gastric cancer and normal mucosal cells. J Biol Chem. 2019;294(20):8238–58.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Vishwakarma K, Dey R, Bhatt H. Telomerase: a prominent oncological target for development of chemotherapeutic agents. Eur J Med Chem. 2023;249:115121.

    Article  PubMed  CAS  Google Scholar 

  96. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25(3):486–541.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, et al. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther. 2022;7(1):286.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Xie J, Yang Y, Gao Y, He J. Cuproptosis: mechanisms and links with cancers. Mol Cancer. 2023;22(1):46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, et al. The role of necroptosis in cancer biology and therapy. Mol Cancer. 2019;18(1):100.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, et al. Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 2022;19(9):971–92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Shchors K, Massaras A, Hanahan D. Dual targeting of the autophagic regulatory circuitry in gliomas with repurposed drugs elicits cell-lethal autophagy and therapeutic benefit. Cancer Cell. 2015;28(4):456–71.

    Article  PubMed  CAS  Google Scholar 

  103. Bhutia SK, Mukhopadhyay S, Sinha N, Das DN, Panda PK, Patra SK, et al. Autophagy: cancer’s friend or foe? Adv Cancer Res. 2013;118:61–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Smithuis F, Kyaw MK, Phe O, Win T, Aung PP, Oo APP, et al. Effectiveness of five artemisinin combination regimens with or without primaquine in uncomplicated falciparum malaria: an open-label randomised trial. Lancet Infect Dis. 2010;10(10):673–81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Wong YK, Xu C, Kalesh KA, He Y, Lin Q, Wong WF, et al. Artemisinin as an anticancer drug: recent advances in target profiling and mechanisms of action. Med Res Rev. 2017;37(6):1492–517.

    Article  PubMed  CAS  Google Scholar 

  106. Yang N-D, Tan S-H, Ng S, Shi Y, Zhou J, Tan KSW, et al. Artesunate induces cell death in human cancer cells via enhancing lysosomal function and lysosomal degradation of ferritin. J Biol Chem. 2014;289(48):33425–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Chen G-Q, Benthani FA, Wu J, Liang D, Bian Z-X, Jiang X. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 2020;27(1):242–54.

    Article  PubMed  CAS  Google Scholar 

  108. Du J, Wang T, Li Y, Zhou Y, Wang X, Yu X, et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radical Biol Med. 2019;131:356–69.

    Article  CAS  Google Scholar 

  109. Zhang J, Guo L, Zhou X, Dong F, Li L, Cheng Z, et al. Dihydroartemisinin induces endothelial cell anoikis through the activation of the JNK signaling pathway Corrigendum. Oncol Lett. 2016;12(3):1896–900.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a target for anticancer therapy. Nat Rev Clin Oncol. 2011;8(9):528–39.

    Article  PubMed  CAS  Google Scholar 

  111. Wolpin BM, Rubinson DA, Wang X, Chan JA, Cleary JM, Enzinger PC, et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. Oncologist. 2014;19(6):637–8.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Pantziarka P, Vandeborne L, Bouche G. A database of drug repurposing clinical trials in oncology. Front Pharmacol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2021.790952.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Brugmans JP, Thienpont DC, van Wijngaarden I, Vanparijs OF, Schuermans VL, Lauwers HL. Mebendazole in enterobiasis. Radiochemical and pilot clinical study in 1,278 subjects. Jama. 1971;217(3):313–6.

    Article  PubMed  CAS  Google Scholar 

  114. Sasaki J, Ramesh R, Chada S, Gomyo Y, Roth JA, Mukhopadhyay T. The anthelmintic drug mebendazole induces mitotic arrest and apoptosis by depolymerizing tubulin in non-small cell lung cancer cells. Mol Cancer Ther. 2002;1(13):1201–9.

    PubMed  CAS  Google Scholar 

  115. Guerini AE, Triggiani L, Maddalo M, Bonù ML, Frassine F, Baiguini A, et al. Mebendazole as a candidate for drug repurposing in oncology: an extensive review of current literature. Cancers. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers11091284.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Kralova V, Hanušová V, Caltová K, Špaček P, Hochmalová M, Skálová L, Rudolf E. Flubendazole and mebendazole impair migration and epithelial to mesenchymal transition in oral cell lines. Chem Biol Interact. 2018;293:124–32.

    Article  PubMed  CAS  Google Scholar 

  117. Joe NS, Godet I, Milki N, Ain NUI, Oza HH, Riggins GJ, Gilkes DM. Mebendazole prevents distant organ metastases in part by decreasing ITGβ4 expression and cancer stemness. Breast Cancer Res. 2022;24(1):98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Zhang J, Wei W, Zhong Q, Feng K, Yang R, Jiang Q. Budding uninhibited by benzimidazoles 1 promotes cell proliferation, invasion, and epithelial-mesenchymal transition via the Wnt/β-catenin signaling in glioblastoma. Heliyon. 2023;9(6):e16996.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23(1):27–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Sainero-Alcolado L, Liaño-Pons J, Ruiz-Pérez MV, Arsenian-Henriksson M. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ. 2022;29(7):1304–17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Pollak M. Overcoming drug development bottlenecks with repurposing: repurposing biguanides to target energy metabolism for cancer treatment. Nat Med. 2014;20(6):591–3.

    Article  PubMed  CAS  Google Scholar 

  122. Rao Kondapally Seshasai S, Kaptoge S, Thompson A, Di Angelantonio E, Gao P, Sarwar N, et al. Diabetes mellitus, fasting glucose, and risk of cause-specific death. N Engl J Med. 2011;364(9):829–41.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Gallagher EJ, LeRoith D. Obesity and diabetes: the increased risk of cancer and cancer-related mortality. Physiol Rev. 2015;95(3):727–48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Pernicova I, Korbonits M. Metformin—mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol. 2014;10(3):143–56.

    Article  PubMed  CAS  Google Scholar 

  125. Lord SR, Harris AL. Is it still worth pursuing the repurposing of metformin as a cancer therapeutic? Br J Cancer. 2023;128(6):958–66.

    Article  PubMed  PubMed Central  Google Scholar 

  126. De A, Kuppusamy G. Metformin in breast cancer: preclinical and clinical evidence. Curr Probl Cancer. 2020;44(1):100488.

    Article  PubMed  Google Scholar 

  127. Fendt S-M, Bell EL, Keibler MA, Davidson SM, Wirth GJ, Fiske B, et al. Metformin decreases glucose oxidation and increases the dependency of prostate cancer cells on reductive glutamine metabolism. Can Res. 2013;73(14):4429–38.

    Article  CAS  Google Scholar 

  128. Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Can Res. 2007;67(14):6745–52.

    Article  CAS  Google Scholar 

  129. Blandino G, Valerio M, Cioce M, Mori F, Casadei L, Pulito C, et al. Metformin elicits anticancer effects through the sequential modulation of DICER and c-MYC. Nat Commun. 2012;3(1):865.

    Article  PubMed  Google Scholar 

  130. Hald J, Jacobsen E. A drug sensitising the organism to ethyl alcohol. Lancet. 1948;252(6539):1001–4.

    Article  Google Scholar 

  131. Veverka KA, Johnson KL, Mays DC, Lipsky JJ, Naylor S. Inhibition of aldehyde dehydrogenase by disulfiram and its metabolite methyl diethylthiocarbamoyl-sulfoxide. Biochem Pharmacol. 1997;53(4):511–8.

    Article  PubMed  CAS  Google Scholar 

  132. Dorokhov YL, Sheshukova EV, Bialik TE, Komarova TV. Human endogenous formaldehyde as an anticancer metabolite: its oxidation downregulation may be a means of improving therapy. BioEssays. 2018;40(12):1800136.

    Article  Google Scholar 

  133. Falls-Hubert KC, Butler AL, Gui K, Anderson M, Li M, Stolwijk JM, et al. Disulfiram causes selective hypoxic cancer cell toxicity and radio-chemo-sensitization via redox cycling of copper. Free Radical Biol Med. 2020;150:1–11.

    Article  CAS  Google Scholar 

  134. Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 2017;552(7684):194–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells—mechanisms of immune surveillance and escape. Nat Rev Clin Oncol. 2017;14(3):155–67.

    Article  PubMed  CAS  Google Scholar 

  136. Teng MW, Swann JB, Koebel CM, Schreiber RD, Smyth MJ. Immune-mediated dormancy: an equilibrium with cancer. J Leucocyte Biol. 2008;84(4):988–93.

    Article  CAS  Google Scholar 

  137. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3(11):991–8.

    Article  PubMed  CAS  Google Scholar 

  138. Chen DS, Mellman I. Elements of cancer immunity and the cancer–immune set point. Nature. 2017;541(7637):321–30.

    Article  PubMed  CAS  Google Scholar 

  139. Mulder WJM, Ochando J, Joosten LAB, Fayad ZA, Netea MG. Therapeutic targeting of trained immunity. Nat Rev Drug Discov. 2019;18(7):553–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Melero I, Gato M, Shekarian T, Aznar A, Valsesia-Wittmann S, Caux C, et al. Repurposing infectious disease vaccines for intratumoral immunotherapy. J Immunother Cancer. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2019-000443.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Shekarian T, Sivado E, Jallas AC, Depil S, Kielbassa J, Janoueix-Lerosey I, et al. Repurposing rotavirus vaccines for intratumoral immunotherapy can overcome resistance to immune checkpoint blockade. Sci Transl Med. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scitranslmed.aat5025.

    Article  PubMed  Google Scholar 

  142. Aznar MA, Molina C, Teijeira A, Rodriguez I, Azpilikueta A, Garasa S, et al. Repurposing the yellow fever vaccine for intratumoral immunotherapy. EMBO Mol Med. 2020;12(1):e10375.

    Article  PubMed  CAS  Google Scholar 

  143. Mastrangelo G, Rossi CR, Pfahlberg A, Marzia V, Barba A, Baldo M, et al. Is there a relationship between influenza vaccinations and risk of melanoma? A population-based case-control study. Eur J Epidemiol. 2000;16(9):777–82.

    Article  PubMed  CAS  Google Scholar 

  144. Sherr CJ, McCormick F. The RB and p53 pathways in cancer. Cancer Cell. 2002;2(2):103–12.

    Article  PubMed  CAS  Google Scholar 

  145. Van Dyke K, Lantz C, Szustkiewicz C. Quinacrine: mechanisms of antimalarial action. Science. 1970;169(3944):492–3.

    Article  PubMed  Google Scholar 

  146. Gurova KV, Hill JE, Guo C, Prokvolit A, Burdelya LG, Samoylova E, et al. Small molecules that reactivate p53 in renal cell carcinoma reveal a NF-kappaB-dependent mechanism of p53 suppression in tumors. Proc Natl Acad Sci U S A. 2005;102(48):17448–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Batchu RB, Gruzdyn OV, Bryant CS, Qazi AM, Kumar S, Chamala S, et al. Ritonavir-mediated induction of apoptosis in pancreatic cancer occurs via the RB/E2F-1 and AKT pathways. Pharmaceuticals. 2014;7(1):46–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Mandal CC, Ghosh-Choudhury N, Yoneda T, Choudhury GG, Ghosh-Choudhury N. Simvastatin prevents skeletal metastasis of breast cancer by an antagonistic interplay between p53 and CD44. J Biol Chem. 2011;286(13):11314–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Miyajima C, Hayakawa Y, Inoue Y, Nagasaka M, Hayashi H. HMG-CoA reductase inhibitor statins activate the transcriptional activity of p53 by regulating the expression of TAZ. Pharmaceuticals. 2022;15(8):1015.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3(6):401–10.

    Article  PubMed  CAS  Google Scholar 

  151. Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nat Rev Clin Oncol. 2012;9(9):498–509.

    Article  PubMed  CAS  Google Scholar 

  152. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8(8):592–603.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Schein CH. Repurposing approved drugs on the pathway to novel therapies. Med Res Rev. 2020;40(2):586–605.

    Article  PubMed  CAS  Google Scholar 

  154. Eleutherakis-Papaiakovou V, Bamias A, Dimopoulos MA. Thalidomide in cancer medicine. Ann Oncol. 2004;15(8):1151–60.

    Article  PubMed  CAS  Google Scholar 

  155. Yabu T, Tomimoto H, Taguchi Y, Yamaoka S, Igarashi Y, Okazaki T. Thalidomide-induced antiangiogenic action is mediated by ceramide through depletion of VEGF receptors, and is antagonized by sphingosine-1-phosphate. Blood. 2005;106(1):125–34.

    Article  PubMed  CAS  Google Scholar 

  156. Richardson P, Anderson K. Thalidomide and dexamethasone: a new standard of care for initial therapy in multiple myeloma. J Clin Oncol. 2006;24(3):334–6.

    Article  PubMed  Google Scholar 

  157. Como JA, Dismukes WE. Oral azole drugs as systemic antifungal therapy. N Engl J Med. 1994;330(4):263–72.

    Article  PubMed  CAS  Google Scholar 

  158. Tsubamoto H, Ueda T, Inoue K, Sakata K, Shibahara H, Sonoda T. Repurposing itraconazole as an anticancer agent. Oncol Lett. 2017;14(2):1240–6.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Wang SJ, Sun B, Cheng ZX, Zhou HX, Gao Y, Kong R, et al. Dihydroartemisinin inhibits angiogenesis in pancreatic cancer by targeting the NF-κB pathway. Cancer Chemother Pharmacol. 2011;68(6):1421–30.

    Article  PubMed  CAS  Google Scholar 

  160. Bhise NS, Shmueli RB, Sunshine JC, Tzeng SY, Green JJ. Drug delivery strategies for therapeutic angiogenesis and antiangiogenesis. Expert Opin Drug Deliv. 2011;8(4):485–504.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Świtalska M, Filip-Psurska B, Milczarek M, Psurski M, Moszyńska A, Dąbrowska AM, et al. Combined anticancer therapy with imidazoacridinone analogue C-1305 and paclitaxel in human lung and colon cancer xenografts—modulation of tumour angiogenesis. J Cell Mol Med. 2022;26(14):3950–64.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15(11):e493-503.

    Article  PubMed  Google Scholar 

  163. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity. 2019;51(1):27–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Wang J, Wu K, Bai F, Zhai H, Xie H, Du Y, et al. Celecoxib could reverse the hypoxia-induced Angiopoietin-2 upregulation in gastric cancer. Cancer Lett. 2006;242(1):20–7.

    Article  PubMed  CAS  Google Scholar 

  165. Lin XM, Li S, Zhou C, Li RZ, Wang H, Luo W, et al. Cisplatin induces chemoresistance through the PTGS2-mediated anti-apoptosis in gastric cancer. Int J Biochem Cell Biol. 2019;116:105610.

    Article  PubMed  CAS  Google Scholar 

  166. Vergani E, Dugo M, Cossa M, Frigerio S, Di Guardo L, Gallino G, et al. miR-146a-5p impairs melanoma resistance to kinase inhibitors by targeting COX2 and regulating NFkB-mediated inflammatory mediators. Cell Commun Signal. 2020;18(1):156.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Guo Q, Li Q, Wang J, Liu M, Wang Y, Chen Z, et al. A comprehensive evaluation of clinical efficacy and safety of celecoxib in combination with chemotherapy in metastatic or postoperative recurrent gastric cancer patients: A preliminary, three-center, clinical trial study. Medicine. 2019;98(27):e16234.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Gasic GJ, Gasic TB, Galanti N, Johnson T, Murphy S. Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease. Int J Cancer. 1973;11(3):704–18.

    Article  PubMed  CAS  Google Scholar 

  169. Li M, Lotan R, Levin B, Tahara E, Lippman SM, Xu X-C. Aspirin induction of apoptosis in esophageal cancer: a potential for chemoprevention1. Cancer Epidemiol Biomark Prev. 2000;9(6):545–9.

    CAS  Google Scholar 

  170. Kumar D, Rahman H, Tyagi E, Liu T, Li C, Lu R, et al. Aspirin suppresses PGE2 and activates AMP kinase to inhibit melanoma cell motility, pigmentation, and selective tumor growth in vivo. Cancer Prev Res. 2018;11(10):629–42.

    Article  CAS  Google Scholar 

  171. Barnard ME, Beeghly-Fadiel A, Milne GL, Akam EY, Chan AT, Eliassen AH, et al. Urinary PGE-M levels and risk of ovarian cancer. Cancer Epidemiol Biomark Prev. 2019;28(11):1845–52.

    Article  CAS  Google Scholar 

  172. Stegeman I, Bossuyt PM, Yu T, Boyd C, Puhan MA. Aspirin for primary prevention of cardiovascular disease and cancer. A benefit and harm analysis. PLoS One. 2015;10(7):e0127194.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Rothwell PM, Fowkes FG, Belch JF, Ogawa H, Warlow CP, Meade TW. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet. 2011;377(9759):31–41.

    Article  PubMed  CAS  Google Scholar 

  174. Li P, Wu H, Zhang H, Shi Y, Xu J, Ye Y, et al. Aspirin use after diagnosis but not prediagnosis improves established colorectal cancer survival: a meta-analysis. Gut. 2015;64(9):1419–25.

    Article  PubMed  CAS  Google Scholar 

  175. Langley RE, Burdett S, Tierney JF, Cafferty F, Parmar MK, Venning G. Aspirin and cancer: has aspirin been overlooked as an adjuvant therapy? Br J Cancer. 2011;105(8):1107–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood. 2018;131(16):1777–89.

    Article  PubMed  CAS  Google Scholar 

  177. Zhang X, Feng Y, Liu X, Ma J, Li Y, Wang T, Li X. Beyond a chemopreventive reagent, aspirin is a master regulator of the hallmarks of cancer. J Cancer Res Clin Oncol. 2019;145(6):1387–403.

    Article  PubMed  CAS  Google Scholar 

  178. Giovannucci E, Rimm EB, Stampfer MJ, Colditz GA, Ascherio A, Willett WC. Aspirin use and the risk for colorectal cancer and adenoma in male health professionals. Ann Internal Med. 1994;121(4):241–6.

    Article  CAS  Google Scholar 

  179. Holmes MD, Chen WY, Li L, Hertzmark E, Spiegelman D, Hankinson SE. Aspirin intake and survival after breast cancer. J Clin Oncol. 2010;28(9):1467–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Jonsson F, Yin L, Lundholm C, Smedby KE, Czene K, Pawitan Y. Low-dose aspirin use and cancer characteristics: a population-based cohort study. Br J Cancer. 2013;109(7):1921–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Skriver C, Maltesen T, Dehlendorff C, Skovlund CW, Schmidt M, Sørensen HT, Friis S. Long-term aspirin use and cancer risk: a 20-year cohort study. J Natl Cancer Inst. 2024;116(4):530–8.

    Article  PubMed  CAS  Google Scholar 

  182. McNeil JJ, Gibbs P, Orchard SG, Lockery JE, Bernstein WB, Cao Y, et al. Effect of aspirin on cancer incidence and mortality in older adults. J Natl Cancer Inst. 2021;113(3):258–65.

    Article  PubMed  CAS  Google Scholar 

  183. Regulska K, Regulski M, Karolak B, Michalak M, Murias M, Stanisz B. Beyond the boundaries of cardiology: still untapped anticancer properties of the cardiovascular system-related drugs. Pharmacol Res. 2019;147:104326.

    Article  PubMed  CAS  Google Scholar 

  184. Shah P, Garris R, Abboud R, Vasudev R, Patel H, Doshi R, et al. Meta-analysis comparing usefulness of beta blockers to preserve left ventricular function during anthracycline therapy. Am J Cardiol. 2019;124(5):789–94.

    Article  PubMed  Google Scholar 

  185. Spini A, Roberto G, Gini R, Bartolini C, Bazzani L, Donnini S, et al. Evidence of β-blockers drug repurposing for the treatment of triple negative breast cancer: a systematic review. Neoplasma. 2019;66(6):963–70.

    Article  PubMed  CAS  Google Scholar 

  186. Hagen R, Ghareeb E, Jalali O, Zinn Z. Infantile hemangiomas: what have we learned from propranolol? Curr Opin Pediatr. 2018;30(4):499–504.

    Article  PubMed  CAS  Google Scholar 

  187. Wagner MJ, Cranmer LD, Loggers ET, Pollack SM. Propranolol for the treatment of vascular sarcomas. J Exp Pharmacol. 2018;10:51–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Breckenridge A, Jacob R. Overcoming the legal and regulatory barriers to drug repurposing. Nat Rev Drug Discovery. 2019;18(1):1–2.

    Article  PubMed  CAS  Google Scholar 

  189. Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme V, Vikas P. The repurposing drugs in oncology (ReDO) project. Ecancermedicalscience. 2014;8(442):2014.

    Google Scholar 

  190. Cha Y, Erez T, Reynolds I, Kumar D, Ross J, Koytiger G, et al. Drug repurposing from the perspective of pharmaceutical companies. Br J Pharmacol. 2018;175(2):168–80.

    Article  PubMed  CAS  Google Scholar 

  191. Heled Y. Patents v. Statutory exclusivities in biological pharmaceuticals-do we really need both. Mich Telecomm Tech L Rev. 2011;18:419.

    Google Scholar 

  192. Halabi S. The drug repurposing ecosystem: intellectual property incentives, market exclusivity, and the future of “new” medicines. Yale J Law Technol. 2018;20:1.

    Google Scholar 

  193. Smith RB. Repositioned drugs: integrating intellectual property and regulatory strategies. Drug Discov Today: Ther Strat. 2011;8(3–4):131–7.

    Google Scholar 

  194. Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021–6.

    Article  PubMed  Google Scholar 

  195. Wieder R, Adam N. Drug repositioning for cancer in the era of AI, big omics, and real-world data. Crit Rev Oncol Hematol. 2022;175:103730.

    Article  PubMed  Google Scholar 

  196. Verbaanderd C, Meheus L, Huys I, Pantziarka P. Repurposing drugs in oncology: next steps. Trends in cancer. 2017;3(8):543–6.

    Article  PubMed  CAS  Google Scholar 

  197. Stewart AK, Jacobus S, Fonseca R, Weiss M, Callander NS, Chanan-Khan AA, Rajkumar SV. Melphalan, prednisone, and thalidomide vs melphalan, prednisone, and lenalidomide (ECOG E1A06) in untreated multiple myeloma. Blood. 2015;126(11):1294–301.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Zweegman S, van der Holt B, Mellqvist UH, Salomo M, Bos GM, Levin MD, et al. Melphalan, prednisone, and lenalidomide versus melphalan, prednisone, and thalidomide in untreated multiple myeloma. Blood. 2016;127(9):1109–16.

    Article  PubMed  CAS  Google Scholar 

  199. Tran AA, Prasad V. Drug repurposing for cancer treatments: a well-intentioned, but misguided strategy. Lancet Oncol. 2020;21(9):1134–6.

    Article  PubMed  CAS  Google Scholar 

  200. Tyrer F, Bhaskaran K, Rutherford MJ. Immortal time bias for life-long conditions in retrospective observational studies using electronic health records. BMC Med Res Methodol. 2022;22(1):86.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Suissa S, Azoulay L. Metformin and the risk of cancer: time-related biases in observational studies. Diabetes Care. 2012;35(12):2665–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Hammer GP, du Prel J-B, Blettner M. Avoiding bias in observational studies: part 8 in a series of articles on evaluation of scientific publications. Dtsch Arztebl Int. 2009;106(41):664.

    PubMed  PubMed Central  Google Scholar 

  203. Devita VT Jr, Young RC, Canellos GP. Combination versus single agent chemotherapy: a review of the basis for selection of drug treatment of cancer. Cancer. 1975;35(1):98–110.

    Article  PubMed  Google Scholar 

  204. Day D, Siu LL. Approaches to modernize the combination drug development paradigm. Genome Med. 2016;8:1–14.

    Article  Google Scholar 

  205. Michel MC, Staskin D. Study designs for evaluation of combination treatment: focus on individual patient benefit. Biomedicines. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines10020270.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Lee SJC, Murphy CC, Geiger AM, Gerber DE, Cox JV, Nair R, Skinner CS. Conceptual model for accrual to cancer clinical trials. J Clin Oncol. 2019;37(23):1993.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Gonzalez-Cao M, Boada A, Teixidó C, Fernandez-Figueras MT, Mayo C, Tresserra F, et al. Fatal gastrointestinal toxicity with ipilimumab after BRAF/MEK inhibitor combination in a melanoma patient achieving pathological complete response. Oncotarget. 2016;7(35):56619.

    Article  PubMed  PubMed Central  Google Scholar 

  208. Chapman PB, Einhorn LH, Meyers ML, Saxman S, Destro AN, Panageas KS, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 1999;17(9):2745.

    Article  PubMed  CAS  Google Scholar 

  209. Schein CH. Repurposing approved drugs on the pathway to novel therapies. Med Res Rev. 2020;40(2):586–605.

    Article  PubMed  CAS  Google Scholar 

  210. Bartoszewski R, Sikorski AF. Editorial focus: understanding off-target effects as the key to successful RNAi therapy. Cell Mol Biol Lett. 2019;24(1):69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Rationalizing combination therapies. Nature Medicine. 2017;23(10):1113.

  212. Maziarz M, Stencel A. The failure of drug repurposing for COVID-19 as an effect of excessive hypothesis testing and weak mechanistic evidence. Hist Philos Life Sci. 2022;44(4):47.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Nguyen N, Jennen D, Kleinjans J. Omics technologies to understand drug toxicity mechanisms. Drug Discov Today. 2022;27(11):103348.

    Article  PubMed  CAS  Google Scholar 

  214. Verbist B, Klambauer G, Vervoort L, Talloen W, Shkedy Z, Thas O, et al. Using transcriptomics to guide lead optimization in drug discovery projects: lessons learned from the QSTAR project. Drug Discov Today. 2015;20(5):505–13.

    Article  PubMed  CAS  Google Scholar 

  215. Bartoszewska S, Króliczewski J, Crossman DK, Pogorzelska A, Bagiński M, Collawn JF, Bartoszewski R. Triazoloacridone C-1305 impairs XBP1 splicing by acting as a potential IRE1α endoribonuclease inhibitor. Cell Mol Biol Lett. 2021;26(1):11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Króliczewski J, Bartoszewska S, Dudkowska M, Janiszewska D, Biernatowska A, Crossman DK, et al. Utilizing genome-wide mRNA profiling to identify the cytotoxic chemotherapeutic mechanism of triazoloacridone C-1305 as direct microtubule stabilization. Cancers. 2020;12(4):864.

    Article  PubMed  PubMed Central  Google Scholar 

  217. Pruteanu L-L, Bender A. Using transcriptomics and cell morphology data in drug discovery: the long road to practice. ACS Med Chem Lett. 2023;14(4):386–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  218. Meissner F, Geddes-McAlister J, Mann M, Bantscheff M. The emerging role of mass spectrometry-based proteomics in drug discovery. Nat Rev Drug Discov. 2022;21(9):637–54.

    Article  PubMed  CAS  Google Scholar 

  219. Page MJ, Amess B, Rohlff C, Stubberfield C, Parekh R. Proteomics: a major new technology for the drug discovery process. Drug Discov Today. 1999;4(2):55–62.

    Article  PubMed  CAS  Google Scholar 

  220. Frantzi M, Latosinska A, Mischak H. Proteomics in drug development: the dawn of a new era? Proteomics Clin Appl. 2019;13(2):e1800087.

    Article  PubMed  Google Scholar 

  221. Pang H, Hu Z. Metabolomics in drug research and development: the recent advances in technologies and applications. Acta Pharm Sin B. 2023;13(8):3238–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  222. Wishart DS. Emerging applications of metabolomics in drug discovery and precision medicine. Nat Rev Drug Discov. 2016;15(7):473–84.

    Article  PubMed  CAS  Google Scholar 

  223. Van de Sande B, Lee JS, Mutasa-Gottgens E, Naughton B, Bacon W, Manning J, et al. Applications of single-cell RNA sequencing in drug discovery and development. Nat Rev Drug Discov. 2023;22(6):496–520.

    Article  PubMed  PubMed Central  Google Scholar 

  224. Huang Y, Dong D, Zhang W, Wang R, Lin Y-C-D, Zuo H, et al. DrugRepoBank: a comprehensive database and discovery platform for accelerating drug repositioning. Database. 2024;2024:baae051.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  225. Masoudi-Sobhanzadeh Y, Omidi Y, Amanlou M, Masoudi-Nejad A. Drug databases and their contributions to drug repurposing. Genomics. 2020;112(2):1087–95.

    Article  PubMed  CAS  Google Scholar 

  226. Corsello SM, Nagari RT, Spangler RD, Rossen J, Kocak M, Bryan JG, et al. Discovering the anticancer potential of non-oncology drugs by systematic viability profiling. Nat Cancer. 2020;1(2):235–48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  227. Ianevski A, Kushnir A, Nader K, Miihkinen M, Xhaard H, Aittokallio T, Tanoli Z. RepurposeDrugs: an interactive web-portal and predictive platform for repurposing mono- and combination therapies. Brief Bioinform. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bib/bbae328.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Kang BH, Plescia J, Song HY, Meli M, Colombo G, Beebe K, et al. Combinatorial drug design targeting multiple cancer signaling networks controlled by mitochondrial Hsp90. J Clin Invest. 2009;119(3):454–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Czech T, Lalani R, Oyewumi MO. Delivery systems as vital tools in drug repurposing. AAPS PharmSciTech. 2019;20(3):116.

    Article  PubMed  CAS  Google Scholar 

  230. Séguy L, Groo A-C, Malzert-Fréon A. How nano-engineered delivery systems can help marketed and repurposed drugs in Alzheimer’s disease treatment? Drug Discov Today. 2022;27(6):1575–89.

    Article  PubMed  Google Scholar 

  231. Weir SJ, DeGennaro LJ, Austin CP. Repurposing approved and abandoned drugs for the treatment and prevention of cancer through public–private partnership. Can Res. 2012;72(5):1055–8.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to apologize to all those who have made significant and important contributions to our understanding of this research area that are not referenced in this manuscript.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote and revised the paper.

Corresponding author

Correspondence to Rafal Bartoszewski.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing financial interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Czechowicz, P., Więch-Walów, A., Sławski, J. et al. Old drugs, new challenges: reassigning drugs for cancer therapies. Cell Mol Biol Lett 30, 27 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s11658-025-00710-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s11658-025-00710-0

Keywords