Next Article in Journal
Tumor Size and Oncological Outcomes in Patients with Early Cervical Cancer Treated by Fertility Preservation Surgery: A Multicenter Retrospective Cohort Study
Next Article in Special Issue
The Role of Indocyanine Green Fluorescence in Rectal Cancer Robotic Surgery: A Narrative Review
Previous Article in Journal
Molecular Mechanisms and Current Treatment Options for Cancer Cachexia
Previous Article in Special Issue
In Vivo and In Vitro Enhanced Tumoricidal Effects of Metformin, Active Vitamin D3, and 5-Fluorouracil Triple Therapy against Colon Cancer by Modulating the PI3K/Akt/PTEN/mTOR Network
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overcoming Therapy Resistance in Colon Cancer by Drug Repurposing

1
Faculty of Medicine, Lebanese University, Beirut 1003, Lebanon
2
Internal Medicine Residency Program, Department of Medicine, Boston University Medical Center, Boston, MA 02218, USA
3
Department of Research, Foundation for Clinic, Public Health, and Epidemiological Research of Venezuela (FISPEVEN), Caracas 1050, Venezuela
4
Baylor St. Luke’s Medical Center, Houston, TX 770030, USA
5
The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
6
Head of Hematology-Oncology Division, Sahel General Hospital, Beirut 1002, Lebanon
7
President of the Lebanese Society of Medical Oncology (LSMO), Beirut 1003, Lebanon
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(9), 2105; https://doi.org/10.3390/cancers14092105
Submission received: 16 March 2022 / Revised: 19 April 2022 / Accepted: 21 April 2022 / Published: 23 April 2022
(This article belongs to the Collection New Treatment for Colorectal Cancer)

Abstract

:

Simple Summary

Despite improvements in standardized screening methods and the development of promising therapies for colorectal cancer (CRC), survival rates are still low. Drug repurposing offers an affordable solution to achieve new indications for previously approved drugs that could play a protagonist or adjuvant role in the treatment of CRC. In this review, we summarize the current data supporting drug repurposing as a feasible option for patients with CRC.

Abstract

Colorectal cancer (CRC) is the third most common cancer in the world. Despite improvement in standardized screening methods and the development of promising therapies, the 5-year survival rates are as low as 10% in the metastatic setting. The increasing life expectancy of the general population, higher rates of obesity, poor diet, and comorbidities contribute to the increasing trends in incidence. Drug repurposing offers an affordable solution to achieve new indications for previously approved drugs that could play a protagonist or adjuvant role in the treatment of CRC with the advantage of treating underlying comorbidities and decreasing chemotherapy toxicity. This review elaborates on the current data that supports drug repurposing as a feasible option for patients with CRC with a focus on the evidence and mechanism of action promising repurposed candidates that are widely used, including but not limited to anti-malarial, anti-helminthic, anti-inflammatory, anti-hypertensive, anti-hyperlipidemic, and anti-diabetic agents.

Graphical Abstract

1. Introduction

Colorectal cancer (CRC) is the third most prevalent cancer in the world, with more than 100,000 new cases and 50,000 deaths occurring in the United States during 2021 [1]. It is the third most common cancer in both sexes and the second most common cancer-related mortality cause [2]. It is projected that there will be an overall doubling of CRC cases in the following decades [3], with an estimated worldwide increase to 2.5 million new cases in 2035 [4]. In countries with a high human development index, the incidence and mortality rates of CRC have decreased predominantly in older adults, while in lower- and middle-income countries, mortality is increasing [2]. There is a growing incidence of CRC in younger groups, likely attributed to a high prevalence of certain risk factors such as poor diet, low physical activity, and higher rates of obesity [2].
Most CRCs arise from neoplastic polyps as stem cells progressively acquire genetic and epigenetic alterations [4]. Chromosomal instability, mismatch repair deficiency, and CpG hypermethylation underline the pathogenesis of CRC [5]. Significant molecular heterogeneity with altered mismatch repair mechanisms leading to microsatellite instability in BRAF-mutated sessile serrated adenomatous polyps and adenocarcinomas increase after 85 years of age [6]. Sporadic CRC accounts for 70% of new cases, usually following a specific succession of mutations in the adenomatous polyposis coli (APC) gene, followed by KRAS, TP53, and DCC mutations [7]. Familial cases correspond to approximately 25% of cases, and 5% occur in well-defined hereditary CRC syndromes such as familial adenomatous polyposis and Lynch syndrome [7].
The stage of the CRC at the time of diagnosis is crucial in determining survival. While the 5-year survival is approximately 90% for patients with stage I disease, in stage IV it decreases to less than 10%, highlighting the importance of early detection [8]. For localized early-stage CRC, surgery is the standard therapy [5]. In patients with stage II CRC with a high risk of recurrence after surgery or stage III disease (lymph node metastases), adjuvant chemotherapy using oxaliplatin and irinotecan in addition to 5-fluorouracil (5FU) or Capecitabine is used [9]. Targeted therapies, including monocolonal antibodies against VEGF (bevacizumab) or EGFR (cetuximab or panitumumab) are used in the metastatic setting in combination with chemotherapy depending on RAS mutational status and tumor location (left vs. right) [9,10]. In addition, immunotherapy has proven to be an effective treatment modality in the setting of microsatellite instability (MSI) high CRC [10]. For patients with unresectable lesions, treatment is a combination of maximum tumoral cytoreduction and chemotherapy [11]. Despite the advances in treatment, advanced disease remains to be associated with poor survival and resistance to cytotoxic and targeted chemotherapies is still occurring [12].

2. Repurposing Approved Drugs in Colon Cancer

Drug repurposing or repositioning involves using approved drugs for conditions different from their original indication [13,14,15]. Several drugs have acquired additional use in the past years and have been reintroduced into practice fueled by this phenomenon. For instance, thalidomide, discontinued from its original use as an antiemetic, is currently used for multiple myeloma [16] and moderate to severe erythema nodosum leprosum [17]. Another example is Sildenafil which preserves both its primary indication for erectile dysfunction [18] and repurposed indication as a treatment option for idiopathic pulmonary hypertension [19].
Drug repurposing has regained a significant role as a convenient, fast, and relatively safe drug development strategy. New drug development usually takes around 10–15 years on average [20], with a success rate reported from 2 to 10% [21,22]. According to the U.S. Food and Drug Administration (FDA), as of 2018, the compound percentage of drugs reaching stage 4 clinical trials was around 6% [23]. Drug repurposing offers significantly shorter development times and lower investments described as 160 million times lower, particularly costs regarding safety testing, molecular characterization, safety profiling, and initial marketing. It leverages known genetic, pharmacodynamic, pharmacokinetic, and adverse effect profiles, usually bypassing stage 1 clinical trials [24]. Therefore, this approach represents a more cost-efficient, expedited, and less risky strategy than traditional drug development [21].
Many successful reintroductions and alternative indications second repurposing as a feasible option in many areas of medicine. Aspirin, for example, has acquired a wide range of indications, ranging from acutely therapeutic to prolonged preventative ones [14,25]. The cardiovascular field further illustrates this diversity with the recent supportive evidence of SGLT-2 inhibitors, initially approved for hyperglycemia management, for heart failure management regardless of the patients’ ejection fraction and notwithstanding their diabetes status [26,27,28,29]. Therapeutics for Alzheimer’s disease have been highly reliant on this strategy. Since memantine in 2003, no new drugs had been approved until the FDA granted the recent fast track concession for aducanumab in 2021 [14,30]. As of 2017, approximately 27 FDA-approved drugs were being evaluated for Alzheimer’s disease in stages 1–3 clinical trials [14].
Oncology has also gained benefits from drug repurposing. Estimation is that 5% of the anticancer drugs entering phase 1 clinical trials are eventually approved [31]. Certain calcium channels blockers such as felodipine and amlodipine besylate undermine filopodia stability in cancer cells, decreasing the likelihood of progression, invasion, and metastasis [32]. Metformin, classically an antidiabetic drug, has been described to decrease tumor growth. Although metabolic reprogramming halting oxidative phosphorylation and multi-targeted mTOR inhibition have hypothesized metformin’s antitumoral activity, precise mechanisms remain obscure [21,33,34].
The benefits of drug repurposing are evident after their serendipitous discovery and raise interest in predictive tools to optimize outcomes. Many approaches group together into either experimental or computational models [24,35]. The former usually involves either in vitro analysis measuring affinity and interaction stability, also called binding assays, or combined in vitro/in vivo models using compound libraries to test for cellular lineage changes, known as the phenotypic model. The phenotypic approach aims to reproduce diseases in an experimental cellular environment and relies on known compound libraries to test and characterize cellular responses [24]. Alternatively, known compounds have been assessed using in silico models stemming from structure-based principles: direct molecular docking, inverse molecular docking, and receptor-based pharmacophore searching [36]. Drug-based strategies use established drug information such as pharmacodynamics, biochemical, adverse effect profiles, and genomic data to determine potential alternative uses. Transcriptomics data are especially valuable to depict deviant cellular responses to diverse pathologic states, notably those with solid genetic pathomechanisms. Conversely, knowledge-based strategies use well-characterized molecular disease mechanisms to depict candidates for drug repurposing [35].
Large genetic and disease datasets are becoming publicly available, and computational tools for processing massive data are evolving accordingly. Computational-based drug repurposing uses data mining, machine learning, and network analysis to distill large datasets involving disease-specific transcriptomics, proteomics, drug efficacy, responses, and even clinical variables [35]. This information provides insight into complex biologic processes such as epigenetic regulation in cancer cells. Furthermore, drug repurposing approaches may be used for epigenetic reprogramming of cancer cells to increase susceptibility via differential transcriptome expressions. A study characterized 45 FDA-approved drugs yielding synergistic activity with histone deacetylating agents and methylation inhibitors. Additionally, they characterized 85 FDA-approved medications that antagonized the action of these drug families, thwarting favorable responses in colon cancer cells. Altogether, these findings illustrate the benefits and complexity of drug repurposing to design personalized and highly effective treatment plans that account for previously unknown drug interactions [37] (Table 1).

2.1. Anti-Hypertensives and Anti-Arrhythmic Drugs

Angiotensin I converting enzyme inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs) are commonly used drugs that have life-prolonging effects on patients treated for several diseases including but not limited to hypertension and heart failure [90]. An in vivo study by Kubota et al. suggested that both ACEIs and ARBs suppress colitis-induced CRC by decreasing chronic inflammation and oxidative stress in obese mice [38]. In another study by Kedika et al., patients who had one or more histologically confirmed adenomatous polyps on an index colonoscopy and received lisinopril—an ACEI—had a 41% reduction in the risk of developing similar polyps over the next 3–5 years [39].
Beta blockers (BB) are class II antiarrhythmic drugs used primarily to treat cardiovascular diseases and many other conditions [91]. In a study by Tapioles et al., Nebivolol was shown to selectively inhibit mitochondrial respiration in an HCT-116 colon cancer cell line by decreasing the activity of complex I of the respiratory chain and restraining the growth of colon cancer cells, hinting towards a repurposing potential for this drug in colon cancer [40]. Furthermore, Engineer et al. demonstrated that the combination of ACEI/ARB with BB was associated with increased survival, decreased hospitalization, and decreased tumor progression in advanced CRC [92].

2.2. Nonsteroidal Anti-Inflammatory Drugs

Nonsteroidal anti-inflammatory drugs (NSAIDs) employ their anti-inflammatory, analgesic, and antipyretic properties by inhibiting the cyclooxygenase (COX) enzymes [93]. COX-2 overexpression is a major risk factor for the development of CRC [94]. The therapeutic effect of aspirin in CRC can be explained by inhibition of COX-2 as well as the c-MYC transcription factor [41,42]. Furthermore, aspirin blocks platelet activity which is implicated in cancer metastasis and immune evasion [43]. However, Chan et al. argued that aspirin must be used for more than 10 years to achieve a statistically significant reduction in COX-2 positive cancer [44].
Celecoxib works by selectively and reversibly inhibiting COX-2, and thus acts to decrease inflammation and pain without affecting platelets [95]. Many studies concluded that celecoxib increases radiosensitization of CRC cells [96,97]. Celecoxib also affects p53 by regulating the expression of p21 and CyclinD1 in a COX-2-independent manner, by upregulating BCCIP, increasing radiosensitivity in the HCT116 CRC cell line [45]. A randomized controlled trial by Bertagnolli et al. showed that celecoxib was effective for the secondary prevention of colorectal adenomas and decreased the cumulative incidence of adenomas after 3 years from 60.7% in the placebo arm to 43.2% in patients receiving 200 mg of celecoxib twice daily [46].

2.3. Anti-Hyperlipidemic Drugs

Statins markedly inhibit HMG-CoA reductase, the enzyme that controls the rate-limiting step in the cholesterol synthesis pathway in the liver [98]. Remarkably, in a large study including 1953 patients with CRC and 2015 controls, the use of statins for at least 5 years was associated with a significantly reduced relative risk of developing CRC (odds ratio (OR) = 0.50; 95% confidence interval (CI), 0.40–0.63) [99]. In vivo, lovastatin was shown to restrict cancer progression and metastasis formation by inhibiting MACC1 [47]. In a large meta-analysis including a total of 31 studies and involving more than 1.6 million subjects, statins were shown to have a moderate protective effect against developing CRC [48].

2.4. Anti-Diabetic Drugs

Metformin, an oral anti-diabetic medication used for type 2 diabetes mellitus, is a biguanide drug that increases insulin sensitivity, decreases intestinal absorption of glucose, and decreases its production by the liver [100]. Previous studies have shown a protective effect of metformin in CRC risk and prognosis [101,102]. The current understanding is that metformin inhibits the mammalian target of rapamycin (mTOR) pathway which plays a central role in CRC cell growth and proliferation [49]. Furthermore, metformin downregulates IGF receptor activation through decreasing insulin and insulin growth factor, resulting in decreased proliferation in colorectal neoplasia [103,104]. Inhibition of mTOR is achieved through inhibition of mitochondrial mammalian respiratory chain complex I followed by activation of liver kinase B1 and downstream target Adenosine monophosphate-activated protein kinase (AMPK) [50,51]. Other research has shown that metformin, through modulation of oxidative stress and nuclear factor-κB (NF-κB) inflammatory responses would induce apoptosis in CRC cell lines [52,53]. Metformin may also increase sensitivity of cancer cell lines to chemotherapeutic agents such as 5-Fluorouracil, irinotecan, and paclitaxel [105,106,107].
Dapagliflozin, another oral antihyperglycemic medication used for type 2 diabetes mellitus works by inhibiting the sodium/glucose cotransporter 2 (SGLT2) in the proximal tubules of the kidney [108]. Dapagliflozin decreases the adhesion of CRC cells by affecting cellular interaction with Collagen types I and IV through activating ADAM10, which subsequently causes a loss in the full-length DDR1 [54]. DDR1 binding to Collagens I and IV is necessary to stimulate cell–collagen interactions [109]. Dapagliflozin also decreases colon cell proliferation by increasing Erk phosphorylation in the HCT116 human colon cancer cell line [55]. In a case report by Okada et al., SGLT2 inhibition in combination with the EGFR inhibitor, cetuximab, reduced both tumor size and carcinoembryonic antigen (CEA) levels in CRC with liver metastasis [110].

2.5. Anti-Helminthic Drugs

Mebendazole is a broad spectrum benzimidazole that inhibits microtubule synthesis by blocking tubulin polymerization [111]. Mebendazole has cytotoxic activity against different CRC cell lines such as HCT-116, RKO, HT-29, HT-8, and SW626 [56,112]. Nygren and Larsson reported that mebendazole induced remission of metastatic lesions in a patient with refractory metastatic CRC [113]. Another study carried out on mice with a constitutional mutation in the Adenomatous polyposis coli (APC) gene showed that the combination of mebendazole and sulindac (an NSAID) decreased both the number and size of intestinal microadenomas by inhibiting MYC and COX-2 pathways, angiogenesis, and the release of pro-tumorigenic cytokines [57].
Niclosamide is a salicylamide derivative that acts by uncoupling oxidative phosphorylation and regulating different signaling pathways [114]. Niclosamide downregulated the Wnt/β-catenin cascade, which is aberrantly activated in 80% of sporadic CRC [115], in both in vitro and in vivo studies [58] and resulted in decreased proliferation in multiple human CRC cell lines such as HCT-116, Caco2, and HT-29 [59], possibly via the induction of autophagy [116]. Furthermore, a recent study by Kang et al. demonstrated that niclosamide could be combined with metformin to synergistically inhibit APC-mutant CRC by suppressing Wnt and YAP [117].

2.6. Anti-Retroviral Drugs

Tenofovir is a nucleoside antiretroviral drug that acts by inhibiting the reverse transcriptase enzyme [118]. Tenofovir also inhibits the activity of human telomerase [119], a crucial enzyme for tumorigenesis and cancer proliferation, whose inhibition represents a promising therapeutic strategy in cancer treatment [120,121]. Sherif et al. demonstrated that rats receiving tenofovir at a dose of 50 mg/kg for 24 weeks had diminished colorectal cell proliferation attributed to decreased Bcl-2 and cyclin D1 expression [60]. Zidovudine, also known as azidothymidine, is another nucleoside reverse transcriptase inhibitor (NRTI) used in the treatment of human immunodeficiency virus (HIV) [122]. Brown et al. demonstrated Zidovudine’s telomerase inhibition activity in the HT-29 colon cancer cell line [61]. Furthermore, Fang et al. showed that the antitumor activity of zidovudine in colon cancer cells is mediated by increased expression of the p53-Puma/Bax/Noxa pathways favoring apoptosis, and activation of the p53-p21 pathway promoting cell cycle arrest [62]. Efavirenz is a non-nucleoside reverse transcriptase inhibitor (NNRTI) used in the treatment of HIV that is selectively cytotoxic to different tumor cell lines, including colorectal carcinoma, by activating the phosphorylation of p53 [63].
Protease inhibitors (PI) are also drugs that suppress the action of HIV proteases to inhibit viral growth, infectivity, and replication [123]. Indinavir and Saquinavir are PI that suppress the growth of human tumor cells by blocking angiogenesis and matrix metalloproteinases to inhibit tumor invasion and progression. [64]. Furthermore, Mühl et al. reported that Ritonavir synergizes with butyrate to induce apoptosis of CRC cells [66]. The anticancer effect of Ritonavir is most likely due to the inhibition of proteolytic degradation, which causes the accumulation of p21 [68], the decreased production of TNF-α, IL-6, IL-8, and VEGF [67], and the increased expression of anti-inflammatory heme oxygenase-1 [66].
Integrase inhibitors are the latest class of antiretroviral drugs which were approved for HIV therapy due to their efficacy, tolerability, and safety [124]. Raltegravir is an integrase inhibitor that inhibits the Fascin-1-dependent invasion of colorectal tumor cells in vitro and in vivo [69]. Fascin-1 is an actin cross-linking protein whose elevated expression is associated with aggressive clinical progression, dismal prognosis, increased recurrence, and worse survival outcomes in patients with CRC [125,126].

2.7. Anti-Microbials

Other anti-microbial drugs have been investigated for repurposing in colon cancer treatment including doxycycline, a semi-synthetic antibiotic derivative of tetracycline used in the treatment of a wide variety of infections [127]. Doxycycline has also been shown to inhibit matrix metalloproteinases [128]. Onoda et al. demonstrated that a combination therapy consisting of doxycycline and a COX-2 inhibitor suppressed colon cancer cell proliferation and invasion [71]. Doxycycline reportedly induced apoptosis in a dose-dependent manner through activation of caspases, release of cytochrome C, and translocation of Bax [70].
Another antibiotic with potential in cancer therapeutics is clarithromycin. Clarithromycin is a potent inhibitor of tumor-induced angiogenesis [73] showing increased efficacy when combined with approved anticancer drugs [72,75,129]. It is also implicated in attenuating autophagy in myeloma cells [130]. Targeting autophagy is considered a promising strategy for colon cancer therapy [131,132]. In a study by Petroni et al., clarithromycin was indeed shown to modulate autophagy in human CRC cells and inhibited the growth of tumors by targeting hERG1 [74].
The inhibition of autophagy as a mechanism of anticancer activity is also shared by azithromycin, another macrolide antibiotic [76,77]. Qiao et al. demonstrated that azithromycin had a synergistic antitumor activity with the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in colon cancer cells. Azithromycin may also suppress autophagy by upregulating the expression of p62 and LC-3B to ultimately induce colon cancer cell death [78].
Gemifloxacin is a fluoroquinolone used in the setting of community-acquired pneumonia and acute exacerbations of chronic bronchitis [133]. Kan et al. demonstrated that gemifloxacin inhibits the migration and invasion of SW620 and LoVol colon cancer cells and downregulates Snail to reduce epithelial-to-mesenchymal transition (EMT). Gemifloxacin also suppresses the NF-κB pathway and cytokine-mediated cell migration and invasion as shown by decreased levels of tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6), IL-8, and vascular endothelial growth factor (VEGF) [79].
Antimalarials are also being considered for the treatment of colon cancer. Artesunate is an antimalarial agent recommended for the treatment of patients with severe Plasmodium falciparum malaria [134]. In a preclinical model of CRC, artesunate was found to suppress inflammation and oxidative stress [81]. Efferth et al. demonstrated a cytotoxic action of artesunate on tumor cells via both p53-dependent and -independent pathways [135] implicated in downregulation of β-catenin [80]. Mefloquine, another antimalarial drug, was found to induce growth arrest and apoptosis of CRC cells in mice via inhibition of the tumor NF-κB signaling pathway [82].

2.8. Others

Drugs used for neurological conditions are increasingly being considered as therapeutic options in cancer patients [136,137]. Friedmann et al. demonstrated that valproate, a histone deacetylase inhibitor (HDACi), dose-dependently reduced the viability of adenocarcinoma cell lines, particularly when combined with mitomycin C [138]. Additionally, in a study by Mologni et al., valproate was found to enhance bosutinib cytotoxicity in colon cancer cells [139]. This may be explained by increasing histone hyperacetylation of H3 and H4 to enhance antitumor activity and by relieving HDAC-driven transcriptional repression [83,140].
Fluoxetine is a selective serotonin reuptake inhibitor (SSRI) that is used in the treatment of major depressive disorder [141]. In a study examining murine colitis-associated colon cancer, fluoxetine was found to inhibit NF-κB activation and decrease TNF-α-mediated IκB kinase (IKK) and IκBα phosphorylation; thus suppressing dextran sulfate sodium (DSS)-induced colitis and colitis-associated tumorigenesis [84]. Furthermore, Kannen et al. studied the antiproliferative effects of fluoxetine on HT29 colon cancer cells. Fluoxetine increased the percentage of HT29 cells in the G0/G1 phase of the cell cycle and enhanced the expression of p27 protein. Fluoxetine also suppressed the development of dysplasia and vascularization-related dysplasia in colon tissue, and reduced VEGF expression and the number of cells with angiogenic potential, such as CD133, CD34, and CD31-positive cell clusters [85].
Sirolimus, also known as Rapamycin, is an FDA-approved mTOR inhibitor used in the prophylaxis of renal graft rejection [142]. Mussin et al. demonstrated that a combination of sirolimus and metformin synergistically inhibits colon tumor growth both in vitro and in vivo [86]. He et al. revealed that mTOR inhibitors induce apoptosis in colon cancer cells via CHOP-dependent DR5 induction of 4E-BP1 dephosphorylation resulting in decreased tumor proliferation, angiogenesis, and invasion [88]. In addition, Wang et al. demonstrated that sirolimus suppresses the FBXW7-loss-driven EMT through its mTOR inhibition activity, thereby decreasing CRC cell migration and invasion [87].
Finally, it is worth noting that probiotics have also been considered for the treatment of CRC. For instance, Hu et al. found that butyrate decreased the transcription of the pro-proliferative miR-92a in human CRC cells [89]. Furthermore, butyrate can also induce apoptosis of DLD-1 colon cancer cells by synergizing with ritonavir [66].

3. Clinical Trials on Drug Repurposing in Colon Cancer

Despite the heterogeneity of potential drugs that can be repurposed, only a few collectively form the most promising treatments for CRC and have been incorporated in clinical trials. A brief overview of the mechanism of action of these drugs is summarized in Figure 1.
Aspirin is the most represented drug in current clinical trials investigating candidate drugs for repurposing in the treatment of CRC (Table 2). One phase 3 clinical trial involves the use of aspirin as an adjuvant component in stages II and III PIK3CA-mutated colon cancer patients. The trial aims to determine whether the daily consumption of 100 mg of aspirin is effective in reducing recurrence and improving survival compared to placebo (ClinicalTrials.gov; NCT02467582). Two similar phase 3 clinical trials aim to determine the effect of 80 mg daily adjuvant aspirin on survival in stages II and III colon cancer patients compared to placebo (ClinicalTrials.gov; NCT02301286 and NCT03464305). ASPIK French is another phase 3 clinical trial whose goal is to determine local or distant recurrence or second CRC or death from any cause, whichever occurs first, in patients with surgically resected PI3K-mutated colon cancer taking 100 mg of daily aspirin as compared to placebo (ClinicalTrials.gov; NCT02945033). Finally, the ASCOLT phase 3 clinical trial is studying the 5-year disease-free and overall survival in patients with Dukes C or high-risk Dukes B CRC taking 200 mg daily aspirin for 3 years (ClinicalTrials.gov; NCT00565708).
NSAIDs are also among the drugs currently under study for repurposing in CRC therapeutics. For instance, the NICHE phase 2 clinical trial involves the administration of celecoxib along with nivolumab and ipilimumab for stages I to III colon cancer in the neoadjuvant setting. The adverse effects will be assessed to determine the regimen’s safety (ClinicalTrials.gov; NCT03026140).
Mebendazole, an anti-helminthic drug, is being tested in a phase 3 clinical trial for its use as an adjuvant component to FOLFOX with Bevacizumab in stage IV CRC patients (ClinicalTrials.gov; NCT03925662). Furthermore, MECORA is a phase 2 clinical trial which aims to examine the effect of metformin in non-diabetic patients with colon cancer where metformin will be administered before and after colon cancer surgery (ClinicalTrials.gov; NCT03359681).
A phase IB clinical trial aims to assess the safety of the antimalarial hydroxychloroquine along with Axitinib and hepatic chemoembolization in subjects with liver dominant metastatic CRC (ClinicalTrials.gov; NCT04873895). Finally, a dose-finding phase 1/2 clinical trial is studying the safety and recommended phase 2 dose (RP2D) of the combination of neratinib and sodium valproate in patients with advanced solid tumors, including RAS-mutated CRC patients (ClinicalTrials.gov; NCT03919292).

4. Conclusions and Future Directions

Despite the advances in oncology, cancer continues to be one of the leading causes of morbidity and mortality worldwide. CRC has the third highest mortality rate of all cancers, with poor survival rates in many groups. Moreover, CRC cases are expected to increase dramatically in the following decades [3]; this is why finding effective treatments for these patients is crucial. Developing new drugs and translating them into clinical practice from phases 1 to 3 is very long and expensive, with only 5% of oncology drugs resulting in FDA approval [33]. For this reason, drug repurposing has gained more interest as a fast and safe way to achieve better outcomes since the pharmacokinetic, pharmacodynamic, and toxicity profiles of these drugs are already established. The investigation of previously approved drugs with other indications for CRC treatment should identify new effective therapies with lower costs and shorter timelines. Many FDA-approved drugs for infectious, cardiovascular, metabolic, and other diseases are currently studied as possible cancer therapies.
It is important to highlight that the heterogeneity of cancer physiology and response to therapy and the multiple resistance mechanisms these cells develop represent an enormous challenge in oncology. The combination of computational frameworks (translational bioinformatics, computational intelligence, and methodological and systems biology) in multidisciplinary teams has successfully sped up clinical trials for repurposing drugs [143]. The emergence of new technologies in this field, such as large-scale multi-omics sequencing, genome-wide positioning systems network (GPSnet) algorithms, and other artificial intelligence algorithms, has helped identify new targets for older drugs [144].
The tools have matured, and novel technological advancements have enabled the identification of drugs that are suitable for repurposing. Pushpakom et al. elegantly highlighted the recent ongoing drug repurposing strategies [145]. In the era of big data, computational-based approaches will be a driving force in this field. These strategies range from mapping drug-binding sites and implicated downstream pathways to developing correlative signatures of transcriptomic and genetic data and their integration with clinical databases [145]. These approaches can help narrow down the list of candidate drugs that can be used for repurposing [146]. Nevertheless, experimental strategies relying on large-scale drug testing using in vitro and in vivo models remain to be an essential bridge to early phase clinical trials [145,147].
However, there are several limitations that might have rendered efforts in drug repurposing unsuccessful. Currently available drugs are approved at specific dosages to treat specific conditions. It is unknown whether these drugs are effective in treating other conditions such as CRC and whether different pharmacodynamic and pharmacokinetic properties are required for their activity in this setting [148]. Clinical trials are crucial to answer these questions; however, funding efforts have remained to be important obstacles as well as regulatory and organizational hurdles [149]. Despite that, not all trials translate into a clinical benefit which has recently been observed during the COVID-19 pandemic where a lot of negative trials have been reported [150]. This may be due to poorly designed trials or due to the fact that in vitro effects observed are simply not always translated into clinical benefit in larger groups of patients [150,151].
In conclusion, the overall impact of repurposing drugs must be studied in terms of survival and other aspects such as toxicity and side effects. Although the success rate is not so high, this is a promising strategy that must be deeply studied to provide new therapies for patients with CRC. Drugs that are currently being tested in clinical trials including but not limited to metformin, mebendazole, aspirin, celecoxib, and valproic acid are promising candidate drugs that might have substantial benefit in the CRC clinic.

Author Contributions

Conceptualization, T.E.Z., M.Y., H.F.B. and N.B.; methodology, T.E.Z., M.Y., D.D.O.-G., M.M., R.N., G.B., H.F.B. and N.B.; validation, H.F.B. and N.B.; data curation, T.E.Z., M.Y., D.D.O.-G., M.M., R.N., G.B. and H.F.B.; writing—original draft preparation, T.E.Z., M.Y., D.D.O.-G., M.M., R.N., G.B. and H.F.B.; writing—review and editing, T.E.Z., H.F.B. and N.B.; visualization, N.B.; supervision, N.B.; project administration, H.F.B.; funding acquisition, H.F.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Figures were created with BioRender.com accessed on 15 April 2022.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Wild, C.; Weiderpass, E.; Stewart, B.W. World Cancer Report: Cancer Research for Cancer Prevention. Cancer Control 2020, 199, 512. [Google Scholar]
  3. Guren, M.G. The global challenge of colorectal cancer. Lancet Gastroenterol. Hepatol. 2019, 4, 894–895. [Google Scholar] [CrossRef] [Green Version]
  4. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
  5. Recio-Boiles, A.C. Colon Cancer; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  6. Itatani, Y.; Kawada, K. Treatment of Elderly Patients with Colorectal Cancer. BioMed Res. Int. 2018, 2018, 2176056. [Google Scholar] [CrossRef] [Green Version]
  7. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [Green Version]
  8. Van der Jeught, K.; Xu, H.C.; Li, Y.J.; Lu, X.B.; Ji, G. Drug resistance and new therapies in colorectal cancer. World J. Gastroenterol. 2018, 24, 3834–3848. [Google Scholar] [CrossRef]
  9. Das, P.K.; Islam, F.; Lam, A.K. The Roles of Cancer Stem Cells and Therapy Resistance in Colorectal Carcinoma. Cells 2020, 9, 1392. [Google Scholar] [CrossRef]
  10. Golshani, G.; Zhang, Y. Advances in immunotherapy for colorectal cancer: A review. Ther. Adv. Gastroenterol. 2020, 13, 1756284820917527. [Google Scholar] [CrossRef]
  11. Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct. Target. Ther. 2020, 5, 22. [Google Scholar] [CrossRef]
  12. Hammond, W.A.; Swaika, A.; Mody, K. Pharmacologic resistance in colorectal cancer: A review. Ther. Adv. Med. Oncol. 2016, 8, 57–84. [Google Scholar] [CrossRef] [Green Version]
  13. Giampieri, R.; Cantini, L.; Giglio, E.; Bittoni, A.; Lanese, A.; Crocetti, S.; Pecci, F.; Copparoni, C.; Meletani, T.; Lenci, E.; et al. Impact of Polypharmacy for Chronic Ailments in Colon Cancer Patients: A Review Focused on Drug Repurposing. Cancers 2020, 12, 2724. [Google Scholar] [CrossRef]
  14. Jourdan, J.P.; Bureau, R.; Rochais, C.; Dallemagne, P. Drug repositioning: A brief overview. J. Pharm. Pharmacol. 2020, 72, 1145–1151. [Google Scholar] [CrossRef] [Green Version]
  15. Nowak-Sliwinska, P.; Scapozza, L.; Ruiz i Altaba, A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 434–454. [Google Scholar] [CrossRef]
  16. Kumar, S.K.; Callander, N.S.; Adekola, K.; Anderson, L.; Baljevic, M.; Campagnaro, E.; Castillo, J.J.; Chandler, J.C.; Costello, C.; Efebera, Y.; et al. Multiple Myeloma, Version 3.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. JNCCN 2020, 18, 1685–1717. [Google Scholar] [CrossRef]
  17. Upputuri, B.; Pallapati, M.S.; Tarwater, P. Thalidomide in the treatment of erythema nodosum leprosum (ENL) in an outpatient setting: A five-year retrospective analysis from a leprosy referral centre in India. PLoS Neglected Trop. Dis. 2020, 14, e0008678. [Google Scholar] [CrossRef]
  18. Fink, H.A.; Mac Donald, R.; Rutks, I.R.; Nelson, D.B.; Wilt, T.J. Sildenafil for male erectile dysfunction: A systematic review and meta-analysis. Arch. Intern. Med. 2002, 162, 1349–1360. [Google Scholar] [CrossRef] [Green Version]
  19. Barnett, C.F.; Machado, R.F. Sildenafil in the treatment of pulmonary hypertension. Vasc. Health Risk Manag. 2006, 2, 411–422. [Google Scholar] [CrossRef]
  20. Institute of Medicine (US) Forum on Drug Discovery, Development, and Translation. Transforming Clinical Research in the United States: Challenges and Opportunities: Workshop Summary; National Academies Press (US): Washington, DC, USA, 2010. [Google Scholar]
  21. Xue, H.; Li, J.; Xie, H.; Wang, Y. Review of Drug Repositioning Approaches and Resources. Int. J. Biol. Sci. 2018, 14, 1232–1244. [Google Scholar] [CrossRef] [Green Version]
  22. U.S. Food and Drug Administration. Step 3: Clinical Research. Available online: https://www.fda.gov/patients/drug-development-process/step-3-clinical-research (accessed on 30 November 2021).
  23. Deotarse, P.J.A.; Baile, M.; Kohle, N.; Kulkarni, A. Drug Repurposing: A Review. Int. J. Pharm. Res. Rev. 2015, 4, 51–58. [Google Scholar]
  24. Parvathaneni, V.; Kulkarni, N.S.; Muth, A.; Gupta, V. Drug repurposing: A promising tool to accelerate the drug discovery process. Drug Discov. Today 2019, 24, 2076–2085. [Google Scholar] [CrossRef]
  25. Moffat, J.G.; Vincent, F.; Lee, J.A.; Eder, J.; Prunotto, M. Opportunities and challenges in phenotypic drug discovery: An industry perspective. Nat. Rev. Drug Discov. 2017, 16, 531–543. [Google Scholar] [CrossRef]
  26. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  27. Zannad, F.; Ferreira, J.P.; Pocock, S.J.; Anker, S.D.; Butler, J.; Filippatos, G.; Brueckmann, M.; Ofstad, A.P.; Pfarr, E.; Jamal, W.; et al. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: A meta-analysis of the EMPEROR-Reduced and DAPA-HF trials. Lancet 2020, 396, 819–829. [Google Scholar] [CrossRef]
  28. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner-La Rocca, H.P.; Choi, D.J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461. [Google Scholar] [CrossRef]
  29. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef] [Green Version]
  30. U.S. Food and Drug Administration. FDA Grants Accelerated Approval for Alzheimer’s Drug. Available online: https://www.fda.gov/news-events/press-announcements/fda-grants-accelerated-approval-alzheimers-drug (accessed on 30 November 2021).
  31. Paul, S.M.; Mytelka, D.S.; Dunwiddie, C.T.; Persinger, C.C.; Munos, B.H.; Lindborg, S.R.; Schacht, A.L. How to improve R&D productivity: The pharmaceutical industry’s grand challenge. Nat. Rev. Drug Discov. 2010, 9, 203–214. [Google Scholar] [CrossRef]
  32. Jacquemet, G.; Baghirov, H.; Georgiadou, M.; Sihto, H. L-type calcium channels regulate filopodia stability and cancer cell invasion downstream of integrin signalling. Nat. Commun. 2016, 7, 13297. [Google Scholar] [CrossRef] [Green Version]
  33. Zhang, Z.; Zhou, L.; Xie, N.; Nice, E.C.; Zhang, T.; Cui, Y.; Huang, C. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 113. [Google Scholar] [CrossRef]
  34. Lord, S.R.; Cheng, W.C.; Liu, D.; Gaude, E.; Haider, S.; Metcalf, T.; Patel, N.; Teoh, E.J.; Gleeson, F.; Bradley, K.; et al. Integrated Pharmacodynamic Analysis Identifies Two Metabolic Adaption Pathways to Metformin in Breast Cancer. Cell Metab. 2018, 28, 679–688.e674. [Google Scholar] [CrossRef] [Green Version]
  35. Jarada, T.N.; Rokne, J.G.; Alhajj, R. A review of computational drug repositioning: Strategies, approaches, opportunities, challenges, and directions. J. Cheminform. 2020, 12, 46. [Google Scholar] [CrossRef]
  36. Liu, X.; Zhu, F.; Ma, X.H.; Shi, Z.; Yang, S.Y.; Wei, Y.Q.; Chen, Y.Z. Predicting targeted polypharmacology for drug repositioning and multi- target drug discovery. Curr. Med. Chem. 2013, 20, 1646–1661. [Google Scholar] [CrossRef]
  37. Raynal, N.J.; Da Costa, E.M.; Lee, J.T.; Gharibyan, V.; Ahmed, S.; Zhang, H.; Sato, T.; Malouf, G.G.; Issa, J.J. Repositioning FDA-Approved Drugs in Combination with Epigenetic Drugs to Reprogram Colon Cancer Epigenome. Mol. Cancer Ther. 2017, 16, 397–407. [Google Scholar] [CrossRef] [Green Version]
  38. Kubota, M.; Shimizu, M.; Sakai, H.; Yasuda, Y.; Ohno, T.; Kochi, T.; Tsurumi, H.; Tanaka, T.; Moriwaki, H. Renin-angiotensin system inhibitors suppress azoxymethane-induced colonic preneoplastic lesions in C57BL/KsJ-db/db obese mice. Biochem. Biophys. Res. Commun. 2011, 410, 108–113. [Google Scholar] [CrossRef]
  39. Kedika, R.; Patel, M.; Pena Sahdala, H.N.; Mahgoub, A.; Cipher, D.; Siddiqui, A.A. Long-term use of angiotensin converting enzyme inhibitors is associated with decreased incidence of advanced adenomatous colon polyps. J. Clin. Gastroenterol. 2011, 45, e12–e16. [Google Scholar] [CrossRef]
  40. Nuevo-Tapioles, C.; Santacatterina, F.; Stamatakis, K.; Núñez de Arenas, C.; Gómez de Cedrón, M.; Formentini, L.; Cuezva, J.M. Coordinate β-adrenergic inhibition of mitochondrial activity and angiogenesis arrest tumor growth. Nat. Commun. 2020, 11, 3606. [Google Scholar] [CrossRef]
  41. Dovizio, M.; Tacconelli, S.; Sostres, C.; Ricciotti, E.; Patrignani, P. Mechanistic and pharmacological issues of aspirin as an anticancer agent. Pharmaceuticals 2012, 5, 1346–1371. [Google Scholar] [CrossRef] [Green Version]
  42. Thun, M.J.; Jacobs, E.J.; Patrono, C. The role of aspirin in cancer prevention. Nat. Rev. Clin. Oncol. 2012, 9, 259–267. [Google Scholar] [CrossRef]
  43. Kopp, H.G.; Placke, T.; Salih, H.R. Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009, 69, 7775–7783. [Google Scholar] [CrossRef] [Green Version]
  44. Chan, A.T.; Ogino, S.; Fuchs, C.S. Aspirin and the risk of colorectal cancer in relation to the expression of COX-2. N. Engl. J. Med. 2007, 356, 2131–2142. [Google Scholar] [CrossRef]
  45. Xu, X.T.; Hu, W.T.; Zhou, J.Y.; Tu, Y. Celecoxib enhances the radiosensitivity of HCT116 cells in a COX-2 independent manner by up-regulating BCCIP. Am. J. Transl. Res. 2017, 9, 1088–1100. [Google Scholar]
  46. Bertagnolli, M.M.; Eagle, C.J.; Zauber, A.G.; Redston, M.; Solomon, S.D.; Kim, K.; Tang, J.; Rosenstein, R.B.; Wittes, J.; Corle, D.; et al. Celecoxib for the prevention of sporadic colorectal adenomas. N. Engl. J. Med. 2006, 355, 873–884. [Google Scholar] [CrossRef]
  47. Juneja, M.; Kobelt, D.; Walther, W.; Voss, C.; Smith, J.; Specker, E.; Neuenschwander, M.; Gohlke, B.O.; Dahlmann, M.; Radetzki, S.; et al. Statin and rottlerin small-molecule inhibitors restrict colon cancer progression and metastasis via MACC1. PLoS Biol. 2017, 15, e2000784. [Google Scholar] [CrossRef] [Green Version]
  48. Qi, J.H.; Wei, J.N.; Zhang, Z.J.; Dong, L.; Zhang, L.; Mao, Y.Y.; Lei, L.J.; Hu, X.Q.; Bai, W.Q. [A Meta-analysis on association between statins and colorectal cancer]. Zhonghua Liu Xing Bing Xue Za Zhi 2021, 42, 343–350. [Google Scholar] [CrossRef]
  49. Slattery, M.L.; Herrick, J.S.; Lundgreen, A.; Fitzpatrick, F.A.; Curtin, K.; Wolff, R.K. Genetic variation in a metabolic signaling pathway and colon and rectal cancer risk: mTOR, PTEN, STK11, RPKAA1, PRKAG2, TSC1, TSC2, PI3K and Akt1. Carcinogenesis 2010, 31, 1604–1611. [Google Scholar] [CrossRef] [Green Version]
  50. Vial, G.; Detaille, D.; Guigas, B. Role of Mitochondria in the Mechanism(s) of Action of Metformin. Front. Endocrinol. 2019, 10, 294. [Google Scholar] [CrossRef] [Green Version]
  51. Zi, F.; Zi, H.; Li, Y.; He, J.; Shi, Q.; Cai, Z. Metformin and cancer: An existing drug for cancer prevention and therapy. Oncol. Lett. 2018, 15, 683–690. [Google Scholar] [CrossRef] [Green Version]
  52. Nguyen, T.T.; Ung, T.T.; Li, S.; Lian, S.; Xia, Y.; Park, S.Y.; Do Jung, Y. Metformin inhibits lithocholic acid-induced interleukin 8 upregulation in colorectal cancer cells by suppressing ROS production and NF-kB activity. Sci. Rep. 2019, 9, 2003. [Google Scholar] [CrossRef] [Green Version]
  53. Saber, M.M.; Galal, M.A.; Ain-Shoka, A.A.; Shouman, S.A. Combination of metformin and 5-aminosalicylic acid cooperates to decrease proliferation and induce apoptosis in colorectal cancer cell lines. BMC Cancer 2016, 16, 126. [Google Scholar] [CrossRef] [Green Version]
  54. Okada, J.; Yamada, E.; Saito, T.; Yokoo, H.; Osaki, A.; Shimoda, Y.; Ozawa, A.; Nakajima, Y.; Pessin, J.E.; Okada, S.; et al. Dapagliflozin Inhibits Cell Adhesion to Collagen I and IV and Increases Ectodomain Proteolytic Cleavage of DDR1 by Increasing ADAM10 Activity. Molecules 2020, 25, 495. [Google Scholar] [CrossRef] [Green Version]
  55. Saito, T.; Okada, S.; Yamada, E.; Shimoda, Y.; Osaki, A.; Tagaya, Y.; Shibusawa, R.; Okada, J.; Yamada, M. Effect of dapagliflozin on colon cancer cell [Rapid Communication]. Endocr. J. 2015, 62, 1133–1137. [Google Scholar] [CrossRef] [Green Version]
  56. Nygren, P.; Fryknäs, M.; Agerup, B.; Larsson, R. Repositioning of the anthelmintic drug mebendazole for the treatment for colon cancer. J. Cancer Res. Clin. Oncol. 2013, 139, 2133–2140. [Google Scholar] [CrossRef] [Green Version]
  57. Williamson, T.; Bai, R.Y.; Staedtke, V.; Huso, D.; Riggins, G.J. Mebendazole and a non-steroidal anti-inflammatory combine to reduce tumor initiation in a colon cancer preclinical model. Oncotarget 2016, 7, 68571–68584. [Google Scholar] [CrossRef] [Green Version]
  58. Sack, U.; Walther, W.; Scudiero, D.; Selby, M.; Kobelt, D.; Lemm, M.; Fichtner, I.; Schlag, P.M.; Shoemaker, R.H.; Stein, U. Novel effect of antihelminthic Niclosamide on S100A4-mediated metastatic progression in colon cancer. J. Natl. Cancer Inst. 2011, 103, 1018–1036. [Google Scholar] [CrossRef] [Green Version]
  59. Osada, T.; Chen, M.; Yang, X.Y.; Spasojevic, I.; Vandeusen, J.B.; Hsu, D.; Clary, B.M.; Clay, T.M.; Chen, W.; Morse, M.A.; et al. Antihelminth compound niclosamide downregulates Wnt signaling and elicits antitumor responses in tumors with activating APC mutations. Cancer Res. 2011, 71, 4172–4182. [Google Scholar] [CrossRef] [Green Version]
  60. Sherif, D.A.; Makled, M.N.; Suddek, G.M. The HIV reverse transcriptase inhibitor Tenofovir suppressed DMH/HFD-induced colorectal cancer in Wistar rats. Fundam. Clin. Pharmacol. 2021, 35, 940–954. [Google Scholar] [CrossRef]
  61. Brown, T.; Sigurdson, E.; Rogatko, A.; Broccoli, D. Telomerase inhibition using azidothymidine in the HT-29 colon cancer cell line. Ann. Surg. Oncol. 2003, 10, 910–915. [Google Scholar] [CrossRef]
  62. Fang, X.; Hu, T.; Yin, H.; Yang, J.; Tang, W.; Hu, S.; Xu, X. Differences in telomerase activity and the effects of AZT in aneuploid and euploid cells in colon cancer. Int. J. Oncol. 2017, 51, 525–532. [Google Scholar] [CrossRef]
  63. Hecht, M.; Harrer, T.; Büttner, M.; Schwegler, M.; Erber, S.; Fietkau, R.; Distel, L.V. Cytotoxic effect of efavirenz is selective against cancer cells and associated with the cannabinoid system. AIDS 2013, 27, 2031–2040. [Google Scholar] [CrossRef]
  64. Toschi, E.; Sgadari, C.; Malavasi, L.; Bacigalupo, I.; Chiozzini, C.; Carlei, D.; Compagnoni, D.; Bellino, S.; Bugarini, R.; Falchi, M.; et al. Human immunodeficiency virus protease inhibitors reduce the growth of human tumors via a proteasome-independent block of angiogenesis and matrix metalloproteinases. Int. J. Cancer 2011, 128, 82–93. [Google Scholar] [CrossRef]
  65. Pajonk, F.; Himmelsbach, J.; Riess, K.; Sommer, A.; McBride, W.H. The human immunodeficiency virus (HIV)-1 protease inhibitor saquinavir inhibits proteasome function and causes apoptosis and radiosensitization in non-HIV-associated human cancer cells. Cancer Res. 2002, 62, 5230–5235. [Google Scholar]
  66. Mühl, H.; Paulukat, J.; Höfler, S.; Hellmuth, M.; Franzen, R.; Pfeilschifter, J. The HIV protease inhibitor ritonavir synergizes with butyrate for induction of apoptotic cell death and mediates expression of heme oxygenase-1 in DLD-1 colon carcinoma cells. Br. J. Pharmacol. 2004, 143, 890–898. [Google Scholar] [CrossRef] [Green Version]
  67. Pati, S.; Pelser, C.B.; Dufraine, J.; Bryant, J.L.; Reitz, M.S., Jr.; Weichold, F.F. Antitumorigenic effects of HIV protease inhibitor ritonavir: Inhibition of Kaposi sarcoma. Blood 2002, 99, 3771–3779. [Google Scholar] [CrossRef] [Green Version]
  68. Gaedicke, S.; Firat-Geier, E.; Constantiniu, O.; Lucchiari-Hartz, M.; Freudenberg, M.; Galanos, C.; Niedermann, G. Antitumor effect of the human immunodeficiency virus protease inhibitor ritonavir: Induction of tumor-cell apoptosis associated with perturbation of proteasomal proteolysis. Cancer Res. 2002, 62, 6901–6908. [Google Scholar]
  69. Alburquerque-González, B.; Bernabé-García, Á.; Bernabé-García, M.; Ruiz-Sanz, J.; López-Calderón, F.F.; Gonnelli, L.; Banci, L.; Peña-García, J.; Luque, I.; Nicolás, F.J.; et al. The FDA-Approved Antiviral Raltegravir Inhibits Fascin1-Dependent Invasion of Colorectal Tumor Cells In Vitro and In Vivo. Cancers 2021, 13, 861. [Google Scholar] [CrossRef]
  70. Onoda, T.; Ono, T.; Dhar, D.K.; Yamanoi, A.; Nagasue, N. Tetracycline analogues (doxycycline and COL-3) induce caspase-dependent and -independent apoptosis in human colon cancer cells. Int. J. Cancer 2006, 118, 1309–1315. [Google Scholar] [CrossRef]
  71. Onoda, T.; Ono, T.; Dhar, D.K.; Yamanoi, A.; Fujii, T.; Nagasue, N. Doxycycline inhibits cell proliferation and invasive potential: Combination therapy with cyclooxygenase-2 inhibitor in human colorectal cancer cells. J. Lab. Clin. Med. 2004, 143, 207–216. [Google Scholar] [CrossRef]
  72. Carella, A.M.; Beltrami, G.; Pica, G.; Carella, A.; Catania, G. Clarithromycin potentiates tyrosine kinase inhibitor treatment in patients with resistant chronic myeloid leukemia. Leuk. Lymphoma 2012, 53, 1409–1411. [Google Scholar] [CrossRef]
  73. Yatsunami, J.; Turuta, N.; Wakamatsu, K.; Hara, N.; Hayashi, S. Clarithromycin is a potent inhibitor of tumor-induced angiogenesis. Res. Exp. Med. 1997, 197, 189–197. [Google Scholar] [CrossRef]
  74. Petroni, G.; Bagni, G.; Iorio, J.; Duranti, C.; Lottini, T.; Stefanini, M.; Kragol, G.; Becchetti, A.; Arcangeli, A. Clarithromycin inhibits autophagy in colorectal cancer by regulating the hERG1 potassium channel interaction with PI3K. Cell Death Dis. 2020, 11, 161. [Google Scholar] [CrossRef] [Green Version]
  75. Schafranek, L.; Leclercq, T.M.; White, D.L.; Hughes, T.P. Clarithromycin enhances dasatinib-induced cell death in chronic myeloid leukemia cells, by inhibition of late stage autophagy. Leuk. Lymphoma 2013, 54, 198–201. [Google Scholar] [CrossRef]
  76. Tanaka, H.; Hino, H.; Moriya, S.; Kazama, H.; Miyazaki, M.; Takano, N.; Hiramoto, M.; Tsukahara, K.; Miyazawa, K. Comparison of autophagy inducibility in various tyrosine kinase inhibitors and their enhanced cytotoxicity via inhibition of autophagy in cancer cells in combined treatment with azithromycin. Biochem. Biophys. Rep. 2020, 22, 100750. [Google Scholar] [CrossRef]
  77. Toriyama, K.; Takano, N.; Kokuba, H.; Kazama, H.; Moriya, S.; Hiramoto, M.; Abe, S.; Miyazawa, K. Azithromycin enhances the cytotoxicity of DNA-damaging drugs via lysosomal membrane permeabilization in lung cancer cells. Cancer Sci. 2021, 112, 3324–3337. [Google Scholar] [CrossRef]
  78. Qiao, X.; Wang, X.; Shang, Y.; Li, Y.; Chen, S.Z. Azithromycin enhances anticancer activity of TRAIL by inhibiting autophagy and up-regulating the protein levels of DR4/5 in colon cancer cells in vitro and in vivo. Cancer Commun. 2018, 38, 43. [Google Scholar] [CrossRef] [Green Version]
  79. Kan, J.Y.; Hsu, Y.L.; Chen, Y.H.; Chen, T.C.; Wang, J.Y.; Kuo, P.L. Gemifloxacin, a fluoroquinolone antimicrobial drug, inhibits migration and invasion of human colon cancer cells. Biomed Res. Int. 2013, 2013, 159786. [Google Scholar] [CrossRef]
  80. Verma, S.; Das, P.; Kumar, V.L. Chemoprevention by artesunate in a preclinical model of colorectal cancer involves down regulation of β-catenin, suppression of angiogenesis, cellular proliferation and induction of apoptosis. Chem. Biol. Interact. 2017, 278, 84–91. [Google Scholar] [CrossRef]
  81. Kumar, V.L.; Verma, S.; Das, P. Artesunate suppresses inflammation and oxidative stress in a rat model of colorectal cancer. Drug Dev. Res. 2019, 80, 1089–1097. [Google Scholar] [CrossRef]
  82. Xu, X.; Wang, J.; Han, K.; Li, S.; Xu, F.; Yang, Y. Antimalarial drug mefloquine inhibits nuclear factor kappa B signaling and induces apoptosis in colorectal cancer cells. Cancer Sci. 2018, 109, 1220–1229. [Google Scholar] [CrossRef] [Green Version]
  83. Patel, M.M.; Patel, B.M. Repurposing of sodium valproate in colon cancer associated with diabetes mellitus: Role of HDAC inhibition. Eur. J. Pharm. Sci. 2018, 121, 188–199. [Google Scholar] [CrossRef]
  84. Koh, S.J.; Kim, J.M.; Kim, I.K.; Kim, N.; Jung, H.C.; Song, I.S.; Kim, J.S. Fluoxetine inhibits NF-κB signaling in intestinal epithelial cells and ameliorates experimental colitis and colitis-associated colon cancer in mice. Am. J. Physiol. Gastrointest Liver Physiol. 2011, 301, G9–G19. [Google Scholar] [CrossRef] [Green Version]
  85. Kannen, V.; Hintzsche, H.; Zanette, D.L.; Silva, W.A.; Garcia, S.B.; Waaga-Gasser, A.M.; Stopper, H. Antiproliferative effects of fluoxetine on colon cancer cells and in a colonic carcinogen mouse model. PLoS ONE 2012, 7, e50043. [Google Scholar] [CrossRef] [Green Version]
  86. Mussin, N.; Oh, S.C.; Lee, K.W.; Park, M.Y.; Seo, S.; Yi, N.J.; Kim, H.; Yoon, K.C.; Ahn, S.W.; Kim, H.S.; et al. Sirolimus and Metformin Synergistically Inhibits Colon Cancer In Vitro and In Vivo. J. Korean Med. Sci. 2017, 32, 1385–1395. [Google Scholar] [CrossRef]
  87. Wang, Y.; Liu, Y.; Lu, J.; Zhang, P.; Xu, Y.; Wang, Z.; Mao, J.H.; Wei, G. Rapamycin inhibits FBXW7 loss-induced epithelial-mesenchymal transition and cancer stem cell-like characteristics in colorectal cancer cells. Biochem. Biophys. Res. Commun. 2013, 434, 352–356. [Google Scholar] [CrossRef] [Green Version]
  88. He, K.; Zheng, X.; Li, M.; Zhang, L.; Yu, J. mTOR inhibitors induce apoptosis in colon cancer cells via CHOP-dependent DR5 induction on 4E-BP1 dephosphorylation. Oncogene 2016, 35, 148–157. [Google Scholar] [CrossRef] [Green Version]
  89. Hu, S.; Liu, L.; Chang, E.B.; Wang, J.Y.; Raufman, J.P. Butyrate inhibits pro-proliferative miR-92a by diminishing c-Myc-induced miR-17-92a cluster transcription in human colon cancer cells. Mol. Cancer 2015, 14, 180. [Google Scholar] [CrossRef] [Green Version]
  90. Hradec, J. Pharmacological therapy for chronic heart failure. Vnitr. Lek. 2018, 64, 853–859. [Google Scholar] [CrossRef]
  91. Do Vale, G.T.; Ceron, C.S.; Gonzaga, N.A.; Simplicio, J.A.; Padovan, J.C. Three Generations of β-blockers: History, Class Differences and Clinical Applicability. Curr. Hypertens. Rev. 2019, 15, 22–31. [Google Scholar] [CrossRef]
  92. Engineer, D.R.; Burney, B.O.; Hayes, T.G.; Garcia, J.M. Exposure to ACEI/ARB and β-Blockers Is Associated with Improved Survival and Decreased Tumor Progression and Hospitalizations in Patients with Advanced Colon Cancer. Transl. Oncol. 2013, 6, 539–545. [Google Scholar] [CrossRef] [Green Version]
  93. Bacchi, S.; Palumbo, P.; Sponta, A.; Coppolino, M.F. Clinical pharmacology of non-steroidal anti-inflammatory drugs: A review. Antiinflamm. Antiallergy Agents Med. Chem. 2012, 11, 52–64. [Google Scholar] [CrossRef]
  94. Sheng, J.; Sun, H.; Yu, F.B.; Li, B.; Zhang, Y.; Zhu, Y.T. The Role of Cyclooxygenase-2 in Colorectal Cancer. Int. J. Med. Sci. 2020, 17, 1095–1101. [Google Scholar] [CrossRef]
  95. McAdam, B.F.; Catella-Lawson, F.; Mardini, I.A.; Kapoor, S.; Lawson, J.A.; FitzGerald, G.A. Systemic biosynthesis of prostacyclin by cyclooxygenase (COX)-2: The human pharmacology of a selective inhibitor of COX-2. Proc. Natl. Acad. Sci. USA 1999, 96, 272–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Pal, I.; Dey, K.K.; Chaurasia, M.; Parida, S.; Das, S.; Rajesh, Y.; Sharma, K.; Chowdhury, T.; Mandal, M. Cooperative effect of BI-69A11 and celecoxib enhances radiosensitization by modulating DNA damage repair in colon carcinoma. Tumour Biol. 2016, 37, 6389–6402. [Google Scholar] [CrossRef] [PubMed]
  97. Dulai, P.S.; Singh, S.; Marquez, E.; Khera, R.; Prokop, L.J.; Limburg, P.J.; Gupta, S.; Murad, M.H. Chemoprevention of colorectal cancer in individuals with previous colorectal neoplasia: Systematic review and network meta-analysis. BMJ 2016, 355, i6188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Bansal, A.B.; Cassagnol, M. HMG-CoA Reductase Inhibitors. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  99. Poynter, J.N.; Gruber, S.B.; Higgins, P.D.; Almog, R.; Bonner, J.D.; Rennert, H.S.; Low, M.; Greenson, J.K.; Rennert, G. Statins and the risk of colorectal cancer. N. Engl. J. Med. 2005, 352, 2184–2192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Buse, J.B.; Wexler, D.J.; Tsapas, A.; Rossing, P.; Mingrone, G.; Mathieu, C.; D’Alessio, D.A.; Davies, M.J. 2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2020, 43, 487–493. [Google Scholar] [CrossRef] [Green Version]
  101. Berkovic, M.C.; Mikulic, D.; Bilic-Curcic, I.; Mrzljak, A. How far along are we in revealing the connection between metformin and colorectal cancer? World J. Gastroenterol. 2021, 27, 1362–1368. [Google Scholar] [CrossRef]
  102. Cunha Júnior, A.D.; Bragagnoli, A.C.; Costa, F.O.; Carvalheira, J.B.C. Repurposing metformin for the treatment of gastrointestinal cancer. World J. Gastroenterol. 2021, 27, 1883–1904. [Google Scholar] [CrossRef]
  103. Pollak, M. Insulin, insulin-like growth factors and neoplasia. Best Pract. Res. Clin. Endocrinol. Metab. 2008, 22, 625–638. [Google Scholar] [CrossRef]
  104. Pollak, M. The insulin and insulin-like growth factor receptor family in neoplasia: An update. Nat. Rev. Cancer 2012, 12, 159–169. [Google Scholar] [CrossRef]
  105. Khader, E.I.; Ismail, W.W.; Mhaidat, N.M.; Alqudah, M.A. Effect of metformin on irinotecan-induced cell cycle arrest in colorectal cancer cell lines HCT116 and SW480. Int. J. Health Sci. 2021, 15, 34–41. [Google Scholar]
  106. Kim, S.H.; Kim, S.C.; Ku, J.L. Metformin increases chemo-sensitivity via gene downregulation encoding DNA replication proteins in 5-Fu resistant colorectal cancer cells. Oncotarget 2017, 8, 56546–56557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Rocha, G.Z.; Dias, M.M.; Ropelle, E.R.; Osório-Costa, F.; Rossato, F.A.; Vercesi, A.E.; Saad, M.J.; Carvalheira, J.B. Metformin amplifies chemotherapy-induced AMPK activation and antitumoral growth. Clin. Cancer Res. 2011, 17, 3993–4005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ganesan, K.; Rana, M.B.M.; Sultan, S. Oral Hypoglycemic Medications. In StatPearls, 2021 January ed.; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  109. Kothiwale, S.; Borza, C.M.; Lowe, E.W.; Pozzi, A.; Meiler, J. Discoidin domain receptor 1 (DDR1) kinase as target for structure-based drug discovery. Drug Discov. Today 2015, 20, 255–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Okada, J.; Matsumoto, S.; Kaira, K.; Saito, T.; Yamada, E.; Yokoo, H.; Katoh, R.; Kusano, M.; Okada, S.; Yamada, M. Sodium Glucose Cotransporter 2 Inhibition Combined With Cetuximab Significantly Reduced Tumor Size and Carcinoembryonic Antigen Level in Colon Cancer Metastatic to Liver. Clin. Colorectal. Cancer 2018, 17, e45–e48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Lacey, E. Mode of action of benzimidazoles. Parasitol. Today 1990, 6, 112–115. [Google Scholar] [CrossRef]
  112. Laudisi, F.; Marônek, M.; Di Grazia, A.; Monteleone, G.; Stolfi, C. Repositioning of Anthelmintic Drugs for the Treatment of Cancers of the Digestive System. Int. J. Mol. Sci. 2020, 21, 4957. [Google Scholar] [CrossRef]
  113. Nygren, P.; Larsson, R. Drug repositioning from bench to bedside: Tumour remission by the antihelmintic drug mebendazole in refractory metastatic colon cancer. Acta Oncol. 2014, 53, 427–428. [Google Scholar] [CrossRef]
  114. Chen, W.; Mook, R.A.; Premont, R.T.; Wang, J. Niclosamide: Beyond an antihelminthic drug. Cell. Signal. 2018, 41, 89–96. [Google Scholar] [CrossRef]
  115. Segditsas, S.; Tomlinson, I. Colorectal cancer and genetic alterations in the Wnt pathway. Oncogene 2006, 25, 7531–7537. [Google Scholar] [CrossRef] [Green Version]
  116. Wang, J.; Ren, X.R.; Piao, H.; Zhao, S.; Osada, T.; Premont, R.T.; Mook, R.A.; Morse, M.A.; Lyerly, H.K.; Chen, W. Niclosamide-induced Wnt signaling inhibition in colorectal cancer is mediated by autophagy. Biochem. J. 2019, 476, 535–546. [Google Scholar] [CrossRef]
  117. Kang, H.E.; Seo, Y.; Yun, J.S.; Song, S.H.; Han, D.; Cho, E.S.; Cho, S.B.; Jeon, Y.; Lee, H.; Kim, H.S.; et al. Metformin and Niclosamide Synergistically Suppress Wnt and YAP in APC-Mutated Colorectal Cancer. Cancers 2021, 13, 3437. [Google Scholar] [CrossRef] [PubMed]
  118. Wassner, C.; Bradley, N.; Lee, Y. A Review and Clinical Understanding of Tenofovir: Tenofovir Disoproxil Fumarate versus Tenofovir Alafenamide. J. Int. Assoc. Provid. AIDS Care 2020, 19, 2325958220919231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Hukezalie, K.R.; Thumati, N.R.; Côté, H.C.; Wong, J.M. In vitro and ex vivo inhibition of human telomerase by anti-HIV nucleoside reverse transcriptase inhibitors (NRTIs) but not by non-NRTIs. PLoS ONE 2012, 7, e47505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Jafri, M.A.; Ansari, S.A.; Alqahtani, M.H.; Shay, J.W. Roles of telomeres and telomerase in cancer, and advances in telomerase-targeted therapies. Genome Med. 2016, 8, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Sanford, S.L.; Welfer, G.A.; Freudenthal, B.D.; Opresko, P.L. Mechanisms of telomerase inhibition by oxidized and therapeutic dNTPs. Nat. Commun. 2020, 11, 5288. [Google Scholar] [CrossRef]
  122. Kinloch-De Loës, S.; Hirschel, B.J.; Hoen, B.; Cooper, D.A.; Tindall, B.; Carr, A.; Saurat, J.H.; Clumeck, N.; Lazzarin, A.; Mathiesen, L. A controlled trial of zidovudine in primary human immunodeficiency virus infection. N. Engl. J. Med. 1995, 333, 408–413. [Google Scholar] [CrossRef]
  123. Flexner, C. HIV-protease inhibitors. N. Engl. J. Med. 1998, 338, 1281–1292. [Google Scholar] [CrossRef]
  124. Blanco, J.L.; Whitlock, G.; Milinkovic, A.; Moyle, G. HIV integrase inhibitors: A new era in the treatment of HIV. Expert Opin. Pharmacother. 2015, 16, 1313–1324. [Google Scholar] [CrossRef]
  125. Shi, S.; Zheng, H.C.; Zhang, Z.G. Roles of Fascin mRNA expression in colorectal cancer: Meta-analysis and bioinformatics analysis. Mol. Clin. Oncol. 2020, 13, 119–128. [Google Scholar] [CrossRef]
  126. Tampakis, A.; Tampaki, E.C.; Nonni, A.; Kostakis, I.D.; Posabella, A.; Kontzoglou, K.; von Flue, M.; Felekouras, E.; Kouraklis, G.; Nikiteas, N. High fascin-1 expression in colorectal cancer identifies patients at high risk for early disease recurrence and associated mortality. BMC Cancer 2021, 21, 153. [Google Scholar] [CrossRef]
  127. Cunha, B.A.; Sibley, C.M.; Ristuccia, A.M. Doxycycline. Ther. Drug Monit. 1982, 4, 115–135. [Google Scholar] [CrossRef] [PubMed]
  128. Jung, J.J.; Razavian, M.; Kim, H.Y.; Ye, Y.; Golestani, R.; Toczek, J.; Zhang, J.; Sadeghi, M.M. Matrix metalloproteinase inhibitor, doxycycline and progression of calcific aortic valve disease in hyperlipidemic mice. Sci. Rep. 2016, 6, 32659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Komatsu, S.; Moriya, S.; Che, X.F.; Yokoyama, T.; Kohno, N.; Miyazawa, K. Combined treatment with SAHA, bortezomib, and clarithromycin for concomitant targeting of aggresome formation and intracellular proteolytic pathways enhances ER stress-mediated cell death in breast cancer cells. Biochem. Biophys. Res. Commun. 2013, 437, 41–47. [Google Scholar] [CrossRef] [PubMed]
  130. Nakamura, M.; Kikukawa, Y.; Takeya, M.; Mitsuya, H.; Hata, H. Clarithromycin attenuates autophagy in myeloma cells. Int. J. Oncol. 2010, 37, 815–820. [Google Scholar] [PubMed]
  131. Mokarram, P.; Albokashy, M.; Zarghooni, M.; Moosavi, M.A.; Sepehri, Z.; Chen, Q.M.; Hudecki, A.; Sargazi, A.; Alizadeh, J.; Moghadam, A.R.; et al. New frontiers in the treatment of colorectal cancer: Autophagy and the unfolded protein response as promising targets. Autophagy 2017, 13, 781–819. [Google Scholar] [CrossRef]
  132. Burada, F.; Nicoli, E.R.; Ciurea, M.E.; Uscatu, D.C.; Ioana, M.; Gheonea, D.I. Autophagy in colorectal cancer: An important switch from physiology to pathology. World J. Gastrointest. Oncol. 2015, 7, 271–284. [Google Scholar] [CrossRef]
  133. Zhanel, G.G.; Fontaine, S.; Adam, H.; Schurek, K.; Mayer, M.; Noreddin, A.M.; Gin, A.S.; Rubinstein, E.; Hoban, D.J. A Review of New Fluoroquinolones: Focus on their Use in Respiratory Tract Infections. Treat. Respir. Med. 2006, 5, 437–465. [Google Scholar] [CrossRef]
  134. Barradell, L.B.; Fitton, A. Artesunate. A review of its pharmacology and therapeutic efficacy in the treatment of malaria. Drugs 1995, 50, 714–741. [Google Scholar] [CrossRef]
  135. Efferth, T.; Sauerbrey, A.; Olbrich, A.; Gebhart, E.; Rauch, P.; Weber, H.O.; Hengstler, J.G.; Halatsch, M.E.; Volm, M.; Tew, K.D.; et al. Molecular modes of action of artesunate in tumor cell lines. Mol. Pharmacol. 2003, 64, 382–394. [Google Scholar] [CrossRef] [Green Version]
  136. Bai, L.; Xu, Z.; Huang, C.; Sui, Y.; Guan, X.; Shi, L. Psychotropic medication utilisation in adult cancer patients in China: A cross-sectional study based on national health insurance database. Lancet Reg. Health West. Pac. 2020, 5, 100060. [Google Scholar] [CrossRef]
  137. Ng, C.G.; Boks, M.P.; Smeets, H.M.; Zainal, N.Z.; de Wit, N.J. Prescription patterns for psychotropic drugs in cancer patients; a large population study in the Netherlands. Psychooncology 2013, 22, 762–767. [Google Scholar] [CrossRef] [PubMed]
  138. Friedmann, I.; Atmaca, A.; Chow, K.U.; Jager, E.; Weidmann, E. Synergistic effects of valproic acid and mitomycin C in adenocarcinoma cell lines and fresh tumor cells of patients with colon cancer. J. Chemother. 2006, 18, 415–420. [Google Scholar] [CrossRef] [PubMed]
  139. Mologni, L.; Cleris, L.; Magistroni, V.; Piazza, R.; Boschelli, F.; Formelli, F.; Gambacorti-Passerini, C. Valproic acid enhances bosutinib cytotoxicity in colon cancer cells. Int. J. Cancer 2009, 124, 1990–1996. [Google Scholar] [CrossRef] [PubMed]
  140. Göttlicher, M.; Minucci, S.; Zhu, P.; Krämer, O.H.; Schimpf, A.; Giavara, S.; Sleeman, J.P.; Lo Coco, F.; Nervi, C.; Pelicci, P.G.; et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J. 2001, 20, 6969–6978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Benfield, P.; Heel, R.C.; Lewis, S.P. Fluoxetine. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in depressive illness. Drugs 1986, 32, 481–508. [Google Scholar] [CrossRef] [PubMed]
  142. Sehgal, S.N. Sirolimus: Its discovery, biological properties, and mechanism of action. Transplant. Proc. 2003, 35, 7S–14S. [Google Scholar] [CrossRef]
  143. Hernández-Lemus, E.; Martínez-García, M. Pathway-Based Drug-Repurposing Schemes in Cancer: The Role of Translational Bioinformatics. Front. Oncol. 2020, 10, 605680. [Google Scholar] [CrossRef]
  144. Fu, L.; Jin, W.; Zhang, J.; Zhu, L.; Lu, J.; Zhen, Y.; Zhang, L.; Ouyang, L.; Liu, B.; Yu, H. Repurposing non-oncology small-molecule drugs to improve cancer therapy: Current situation and future directions. Acta Pharm. Sin. B 2021, 12, 532–557. [Google Scholar] [CrossRef]
  145. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef]
  146. Wu, P.; Feng, Q.; Kerchberger, V.E.; Nelson, S.D.; Chen, Q.; Li, B.; Edwards, T.L.; Cox, N.J.; Phillips, E.J.; Stein, C.M.; et al. Integrating gene expression and clinical data to identify drug repurposing candidates for hyperlipidemia and hypertension. Nat. Commun. 2022, 13, 46. [Google Scholar] [CrossRef]
  147. Zhang, L.; Chen, C.; Fu, J.; Lilley, B.; Berlinicke, C.; Hansen, B.; Ding, D.; Wang, G.; Wang, T.; Shou, D.; et al. Large-scale phenotypic drug screen identifies neuroprotectants in zebrafish and mouse models of retinitis pigmentosa. eLife 2021, 10, e57245. [Google Scholar] [CrossRef] [PubMed]
  148. Oprea, T.I.; Bauman, J.E.; Bologa, C.G.; Buranda, T.; Chigaev, A.; Edwards, B.S.; Jarvik, J.W.; Gresham, H.D.; Haynes, M.K.; Hjelle, B.; et al. Drug Repurposing from an Academic Perspective. Drug Discov. Today Ther. Strateg. 2011, 8, 61–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Neuberger, A.; Oraiopoulos, N.; Drakeman, D.L. Renovation as innovation: Is repurposing the future of drug discovery research? Drug Discov. Today 2019, 24, 1–3. [Google Scholar] [CrossRef] [PubMed]
  150. Martinez, M.A. Lack of Effectiveness of Repurposed Drugs for COVID-19 Treatment. Front. Immunol. 2021, 12, 635371. [Google Scholar] [CrossRef] [PubMed]
  151. Abdullah, M.I.; de Wolf, E.; Jawad, M.J.; Richardson, A. The poor design of clinical trials of statins in oncology may explain their failure—Lessons for drug repurposing. Cancer Treat Rev. 2018, 69, 84–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Mode of action of candidate repurposed drugs being tested in clinical trials for patients with CRC. HDAC: Histone Deacetylase; TNIK: TRAF2 And NCK Interacting Kinase; COX: Cyclo-oxygenase; mTOR: mammalian Target of Rapamycin. Adapted from “Round-Cell Background”, by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates accessed on 15 April 2022.
Figure 1. Mode of action of candidate repurposed drugs being tested in clinical trials for patients with CRC. HDAC: Histone Deacetylase; TNIK: TRAF2 And NCK Interacting Kinase; COX: Cyclo-oxygenase; mTOR: mammalian Target of Rapamycin. Adapted from “Round-Cell Background”, by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates accessed on 15 April 2022.
Cancers 14 02105 g001
Table 1. Summary table of the drugs that have been repurposed to be used in colon cancer.
Table 1. Summary table of the drugs that have been repurposed to be used in colon cancer.
ReferenceDrugOriginal IndicationPossible Mode(s) of ActionEffect(s)
[38,39]ACEIs/ARBsHypertensionDecreased chronic inflammation and oxidative stressReduced risk of adenomatous colon polyps
[40]NebivololHypertension and other indicationsInhibition of mitochondrial respiration by decreasing the activity of Complex I of the respiratory chainSuppressed the growth of colon cancer cells
[41,42,43,44]AspirinAntiplateletInhibition of COX-2, c-MYC transcription factor, and the antiplatelet mechanism of actionDecreased cancer metastasis and immune evasion
[45,46]CelecoxibAnti-inflammatoryEffect on p53 by regulating the expression of p21 and CyclinD1 in a COX-2-independent manner
Upregulation of BCCIP
Increased radiosensitivity in HCT116 cell line
Decreased incidence of adenomatous polyps.
[47,48]LovastatinAntilipidemicInhibition of MACC1Restricted cancer progression and metastasis formation
[49,50,51,52,53]MetforminAntihyperglycemicInhibition of mTOR
Modulation of oxidative stress and nuclear factor-κB inflammatory responses
Apoptosis in CRC cell lines
[54,55]DapagliflozinAntihyperglycemicEffect on cellular interaction with Collagen types I and IV
Increased Erk phosphorylation
Decreased adhesion and proliferation of colon cancer cells
[56,57]MebendazoleAnti-helminthicInhibition of MYCCytotoxic activity against different colon cancer cell lines
[58,59]NiclosamideAnti-helminthicDownregulation of the Wnt/β-catenin cascadeDecreased proliferation in multiple human CRC cell lines
[60]TenofovirAnti-retroviral (anti-HIV drug)Decreased Bcl-2 and cyclin D1 expressionInhibition of proliferation, oxidative stress, and inflammation
[61,62]ZidovudineAnti-retroviral (anti-HIV drug)Increased expression of the p53-Puma/Bax/Noxa pathways
Activation of the p53-p21 pathway
Apoptosis
Cell cycle arrest
[63]EfavirenzAnti-retroviral (anti-HIV drug)Activation of the phosphorylation of p53Cytotoxic activity against different colon cancer cell lines
[64]IndinavirAnti-retroviral (anti-HIV drug)Proteasome-independent block of angiogenesis and matrix metalloproteinasesSuppressed growth
[64,65]SaquinavirAnti-retroviral (anti-HIV drug)Proteasome-independent block of angiogenesis and matrix metalloproteinases
Inhibition of proteolytic degradation and accumulation of p21
Apoptosis
Suppressed growth
[66,67,68]RitonavirAnti-retroviral (anti-HIV drug)Inhibition proteolytic degradation and accumulation of p21
Decreased production of TNF-α, IL-6, IL-8, and VEGF
Increased expression of heme oxygenase-1
Apoptosis
Suppressed angiogenesis
[69]RaltegravirAnti-retroviral (anti-HIV drug)Blockage of fascin-1Suppressed invasion
[70,71]DoxycyclineAntibioticInhibition of matrix metalloproteinases
Activation of caspase-3, -8, and -9
Release of cytochrome c and Bax translocation
Apoptosis
Suppressed proliferation and invasive potential
[72,73,74,75]ClarithromycinAntibioticInhibition of autophagy by targeting hERG1Suppressed angiogenesis
Suppressed growth of colon cancer cells
[76,77,78]AzithromycinAntibioticInhibition of autophagy by upregulating p62 and LC-3BApoptosis
[79]GemifloxacinAntibioticInhibition of NF-κB activation
Inhibition of TNF-α, IL-6, IL-8, and VEGF
Suppressed cell migration and invasion
[80,81]ArtesunateAntimalarialDownregulation of β-cateninApoptosis
Cytotoxicity
[82]MefloquineAntimalarialInhibition of NF-κB activationApoptosis
Growth arrest
[83]ValproateAntipsychoticHistone hyperacetylation
Relief of HDAC-mediated transcriptional repression
Reduced viability
Enhanced cytotoxicity
[84,85]FluoxetineAntidepressantInhibition of NF-κB activation and IKK phosphorylation
Cell-cycle arrest at G0/G1
Enhanced p27 expression
Reduced VEGF expression
Suppressed colitis-associated tumorigenesis
Suppressed dysplasia and angiogenesis
[86,87,88]SirolimusPrevention of kidney transplant rejectionCHOP-dependent DR5 induction on 4E-BP1 dephosphorylation
Suppressed FBXW7 loss-driven EMT
Apoptosis
Decreased angiogenesis
Suppressed proliferation and invasion of colon cancer cells
[89]ButyrateProbioticInhibition of miR-92aSuppressed proliferation of colon cancer cells
Table 2. Examples of some repurposed drugs currently being clinically investigated for the treatment of colon cancer.
Table 2. Examples of some repurposed drugs currently being clinically investigated for the treatment of colon cancer.
Clinical Trial NumberPhaseStatusEstimated Completion DateIntervention/TreatmentPatient PopulationPatients EnrolledPrimary Outcome MeasuresSecondary Outcome Measures
NCT024675823Active, not recruitingJune 2029AspirinStages II and III PIK3CA-mutated CRC previously treated with surgery185DFS after 6 yearsTime to recurrenceOS
Cancer-specific survival
Adverse events
NCT023012863RecruitingSeptember 2022AspirinStages II and III CRC1588OSDFS TTF
NCT034643053RecruitingDecember 2026AspirinStages II and III CRC4005-year OSDFS TTF
NCT029450333RecruitingJuly 2024AspirinPI3K-mutated CRC246Recurrence or second CRC or death, whichever occurs first5-year OS
Adverse events
NCT005657083Active, not recruitingJune 2026AspirinDukes C and high-risk Dukes B CRCs1587DFSOS
NCT030261402RecruitingJanuary 2022Nivolumab + Ipilimumab with or without CelecoxibStages I to III CRC60Incidence of adverse eventsImmune activating capacity of immunotherapyRelapse-free survival
NCT039256623RecruitingDecember 2028FOLFOX + bevacizumab with or without mebendazoleStage IV CRC40ORR-
NCT033596812RecruitingJanuary 2022MetforminCRC48 Ki67 expression on tumor samplesCleaved Caspase-3 expression
Immunoscore
Immunological changes in blood samples
In vitro cell growth
NCT048738951RecruitingNovember 2023Axitinib + hydroxychloroquine Liver-dominant metastatic CRC 25Serious adverse eventsORR in setting of liver metastasis
PFS
OS
NCT039192921/2RecruitingJanuary 2024Neratinib + valproateAdvanced solid tumors including CRC113Recommended phase 2 dose Adverse events
Antitumor effects
PFS
Abbreviations: CRC: colorectal cancer; DFS: disease-free survival; ORR: objective response rate; OS: overall survival; PFS: progression-free survival; TTF: time-to-treatment-failure; FOLFOX: folinic acid + fluorouracil + oxaliplatin.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

El Zarif, T.; Yibirin, M.; De Oliveira-Gomes, D.; Machaalani, M.; Nawfal, R.; Bittar, G.; Bahmad, H.F.; Bitar, N. Overcoming Therapy Resistance in Colon Cancer by Drug Repurposing. Cancers 2022, 14, 2105. https://doi.org/10.3390/cancers14092105

AMA Style

El Zarif T, Yibirin M, De Oliveira-Gomes D, Machaalani M, Nawfal R, Bittar G, Bahmad HF, Bitar N. Overcoming Therapy Resistance in Colon Cancer by Drug Repurposing. Cancers. 2022; 14(9):2105. https://doi.org/10.3390/cancers14092105

Chicago/Turabian Style

El Zarif, Talal, Marcel Yibirin, Diana De Oliveira-Gomes, Marc Machaalani, Rashad Nawfal, Gianfranco Bittar, Hisham F. Bahmad, and Nizar Bitar. 2022. "Overcoming Therapy Resistance in Colon Cancer by Drug Repurposing" Cancers 14, no. 9: 2105. https://doi.org/10.3390/cancers14092105

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop