The first category is classified based on the knowledge and ability of medical practitioners or researchers to organize scientific observations of random events. For instance, ormeloxifene, a clinically approved selective estrogen receptor modulator, exhibits anti-cancer properties in multiple cancers including ovarian, head and neck, and breast cancers. However, Hafeez et al. reported ormeloxifene–mediated inhibition of oncogenic β-catenin signaling and EMT progression in PCa, mainly by repressing N-cadherin, MMPs (MMP2 and MMP3), β-catenin/TCF-4 transcriptional activity, and inducing pGSK3β expression. In addition, treatment with ormeloxifene inhibited tumorigenic, metastatic, and invasive capacity of PCa cells in vitro and reduced prostate tumors in xenograft mouse models
[52]. Naftopidil, a selective adrenoreceptor A1D antagonist, is used for treating lower urinary tract symptoms triggered by benign prostatic hyperplasia
[53]. It is also shown to improve the efficiency of radiotherapy (RT) treatment in PC-3 xenograft models as compared with monotherapy with naftopidil or RT
[54] (
Figure 2).
Nitroxoline, a widely used antibiotic for treating urinary tract infections, inhibits endothelial cell proliferation for different solid cancer types
[76][77]. It also mediates AMPK-dependent inhibition of the mTOR signaling pathway and cyclin D1-Rb-Cdc25A axis suggesting its potential role for therapeutic development against PCa
[78]. Nelfinavir, a human immunodeficiency virus (HIV) protease inhibitor authorized by the FDA, is typically used in therapies for HIV patients
[79]. Studies have identified certain nitroxoline-mediated anti-cancer mechanisms, such as inhibition of (PI3K)/Akt signaling pathway, the proteasome, and HIF-1α which limit angiogenesis, as well as activation of endoplasmic reticulum (ER) stress, autophagy, and apoptosis
[80]. Interestingly, nelfinavir shows promising therapeutic potential for the treatment of CRPC through blocking site-2 protease cleavage
[71].
Second are the drugs that have been evaluated in assays and categorized according to their activity. Itraconazole, the antifungal drug, which inhibits angiogenesis and the Hedgehog signaling pathway, has been tested in phase II clinical trials and found effective in men with metastatic CRPC. In addition, digoxin was repurposed for PCa treatment as it inhibits HIF-1α synthesis and tumor growth and was clinically trialed in patients with recurrent PCa
[81][82]. Clofoctol, an antibiotic used to treat upper respiratory tract infections, is a promising inhibitor of PCa. Wang and colleagues reported that clofoctol reduced the development of PCa cells at clinically feasible doses. It also induced ER stress and activated all three Unfolded Protein Response (UPR) pathways, resulting in indirect suppression of protein translation and subsequent reduction in the amounts of G1/S cyclins, resulting in G1 cell cycle arrest. Furthermore, clofoctol was found to be effective in a PCa xenograft model in animals, making it a promising anti-PCa medication candidate suitable for human testing
[83]. Several fatty acid synthase (FASN) inhibitors, including antifungal agent cerulenin, its synthetic derivative C75, and triclosan have been shown to inhibit cancer cell growth by inducing cell death. However, their evaluation in clinical trials was challenged due to pharmacological limitations
[84][85]. Triclosan (TSC) is an approved bactericide in personal hygiene products that possesses a good safety profile, oral bioavailability, and stability in plasma
[86]. Triclosan was found to be a superior alternative to C75 and orlistat in triggering cell death in PCa cells via the inhibition of FASN. In addition, it induced G0/G1 cell cycle arrest and dose-dependent reduction in total lipid content of PCa cells
[85]. As a result, TCS-mediated suppression of the metabolic oncogene FASN has the potential to be developed as a treatment for advanced PCa.
Other drugs are classified using the in-silico drug repurposing approach. For example, risperidone, an antipsychotic agent, is used for the treatment of central nervous system disorders owing to its binding affinity for dopamine D2 and serotonin 5-HT2 receptors
[87]. On the other hand, 17-β-hydroxysteroid dehydrogenase 10 (17HSD10) is responsible for Alzheimer’s disease pathogenesis and PCa cell survival upon ADT. Since risperidone targets 17HSD10, it is a possible treatment choice for both Alzheimer’s disease and PCa
[88].
3.2.1. Anti-Hypertensives and Anti-Arrhythmic Drugs
Beta-blockers act by blockage of beta-adrenergic receptors in the body and have been traditionally utilized for their anti-hypertensive and anti-arrhythmic properties
[91]. One such beta blocker is the non-selective propranolol, which has been shown to exhibit anticancer properties, encompassing PCa
[55]. Specifically, it has been shown to halt PCa proliferation and induce apoptosis, in synergism with a glucose analog, 2-deoxy-d-glucose (2DG). Propranolol also acts via inhibition of phosphatidic acid phosphatase (PAP), which has been shown to be overexpressed in certain cancers
[92]. After induction by 2DG, propranolol’s blockade of PAPs leads to accumulation of LC3-II and p62, markers of autophagy, thus promoting cancer cell death
[57].
Cardiac glycosides, such as digoxin and ouabain, exert their anti-arrhythmic properties via Na
+/K
+ ATPase pump inhibition, which increases intracellular Ca
2+ levels
[57]. Simspon et al. showed that ouabain sensitized resistant prostate adenocarcinoma cells (PPC-1) to aniokis, the process by which normal cells undergo apoptosis by detaching from the extracellular matrix (ECM) via a caspase-dependent mechanism
[56].
Prazosin is a selective inhibitor of the alpha-1 adrenergic receptor and has been authorized for the treatment of hypertension
[93]. Additionally, it is used to treat a variety of clinical conditions, including benign prostatic hyperplasia, Raynaud’s illness, and congestive heart failure
[93][94][95]. In patient-derived glioblastoma-initiating cells (GICs), prazosin mediated growth inhibition through inhibiting the PKCδ- dependent AKT signaling pathway compared to neural stem cells that lack PKCδ
[96]. Interestingly, prazosin has been shown to display antiproliferative activity, superior to that of other α1-blockers, by inducing G2 checkpoint arrest and subsequent apoptosis in PCa cell lines
[97].
3.2.2. NSAIDS, Anti-Inflammatory Drugs and Aspirin
Non-steroidal anti-inflammatory drugs (NSAIDs) act via inhibition of the cyclooxygenase (COX) pathway, thus limiting prostaglandin (PG) synthesis, and are one of the most widely used drugs for their anti-inflammatory, antipyretic, and analgesic effects
[98]. NSAIDs have been shown to affect the proliferative, apoptotic, resistance, and metastatic potential of many PCa cell lines via both COX-dependent and independent mechanisms. The aforementioned results highlight its potential for use in therapy-resistant PCa
[99]. An example of the COX-independent mechanism is demonstrated in the antitumorigenic effects of statin and aspirin combination on LNCaP cells, which are androgen-sensitive human PCa cells, via reduction in cyclin D1 levels
[58]. Celecoxib, a selective COX-2 inhibitor, blocks Akt phosphorylation and activation, which, in turn, leads to apoptosis in PCa cells
[59]. Another anti-inflammatory agent, the glucocorticoid dexamethasone, inhibited ERG activity, an oncogenic transcription factor, of prostate tumor cells in an in-silico model
[60].
According to clinical studies, patients undergoing long-term NSAID treatment have a decreased chance of acquiring cancer
[100][101]. Indomethacin, an NSAID used for rheumatic disease treatment
[102][103], is a notable antineoplastic agent. Mechanistically, it inhibits Cox-1/2-dependent angiogenesis
[104][105] as well as MAPK pathways
[106]. It also inhibits cancer cell growth via impairing PKC-p38-DRP1 axis-dependent mitochondrial dynamics or downregulating Wnt/β-catenin signaling
[107]. A clinical trial is currently ongoing (
ClinicalTrials.gov; NCT02935205) to study the combined effect of hormone therapy using enzalutamide and indomethacin on PCa by lowering the amount of androgen the body makes and/or blocking the use of androgen by the tumor cells.
3.2.3. Anti-Hyperlipidemic Drugs
Statins act as lipid-lowering agents by inhibiting the HMG-CoA reductase enzyme and thus inhibiting the rate-limiting step cholesterol biosynthesis
[108]. However, preclinical and clinical evidence show that statins exhibit anti-neoplastic activity in various cancers
[109][110]. Ianelli et al. demonstrated the synergistic effects of simvastatin and valproic acid in sensitizing PCa to docletaxel and decreasing resistance rates. This combination method targets the CSCs by inhibiting the action of the Yes-associated protein (YAP) oncogene, a transcriptional regulator implicated in many cancers, including PCa
[61][62]. Zhuang et al. demonstrated that simvastatin acts to promote apoptosis via cholesterol depletion. Specifically, simvastatin acts via depletion of cholesterol-containing lipid rafts of PCa cells, which, in turn, inhibits protein kinase B (also known as AKT) signaling pathway
[111]. Statins also have a role in sensitizing PCa cells to ADT. Kong et al. have shown that simvastatin treatment helps to re-sensitize PCa cell lines that have developed resistance to enzalutamide, an FDA approved anti-androgenic agent for the treatment of CRPC
[63].
In a retrospective cohort study of men who underwent prostate biopsy, statin decreased risk of PCa and resulted in a better prognosis and reduction in PCa volume and PSA levels
[112]. Other studies have indicated that statins reduce PCa-related mortality and risk
[113][114], while others have found no impact
[112]. Inconsistent clinical results make a definite link between statin usage and PCa difficult to establish
[115].
3.2.4. Anti-Diabetic Drugs
Metformin, a member of the biguanide family, acts as an anti-diabetic agent by increasing sensitivity to insulin, inhibiting hepatic production of glucose, and restricting hepatic gluconeogenesis
[116]. Aside from its anti-diabetic properties, metformin has shown the potential to be used for its anti-cancer properties, particularly in PCa. Metformin inhibits the mitochondrial complex I of the electron transport chain, which leads to an increase in adenosine monophosphate-activated protein kinase (AMPK), in turn leading to the inhibition of the mammalian target of rapamycin (mTOR) and activation of tuberous sclerosis complex-2, a tumor suppressor gene
[64]. By decreasing the levels of insulin in circulation, metformin downregulates phosphoinositide-3-kinase (PI3K) axis, a main contributor to cell growth, proliferation, and differentiation
[65]. Furthermore, metformin has been shown to decrease androgen receptor expression, which could establish its role as an adjunct to ADT
[66]. Glipizide, another antidiabetic drug that stimulates beta-cell insulin secretion, has shown tumor suppressive effects of PCa cells in a TRAMP transgenic mouse model. It does so mainly via inhibition of angiogenesis, particularly by targeting HMGIY/ANGPT1 signaling pathway
[67].
Metformin also has significant anti-cancer activity
[117][118][119] in breast, prostate, lung, cervix, ovarian, and CNS cancers, and it is being used in phase 1–4 clinical trials for cancer therapy
[120]. Metformin reduced the risk of PCa diagnosis in comparison to other oral hypoglycemics
[121]. In xenograft models, the combination of metformin and chemotherapy inhibits tumor development and prevents recurrence of PCa cells via inhibiting inflammatory pathways
[122]. In addition, metformin and valproic acid (VPA) synergistically suppressed proliferation in both LNCaP (androgen dependent) and PC-3 (androgen independent) cell lines and enhanced apoptosis in patient-derived prostate tumor explants
[123].
Long-term metformin therapy has also been found to significantly reduce the incidence of breast cancer in women with type 2 diabetes
[124]. By reducing elevated insulin levels, metformin inhibits the growth of tumors expressing insulin receptors
[125][126]. Metformin has also been shown to activate AMPK, a critical energy sensor in cellular metabolism, and to limit cancer cell proliferation via the negative regulation of mTOR needed for tumor survival
[127].
3.2.5. Anti-Helminthic Drugs
Mebendazole is a microtubule inhibiting drug that fights parasitic infections. Docetaxel is a treatment option for metastatic PCa; however, it had a modest effect on increasing the median survival time
[128]. Rushworth et al. showed that mebendazole and docetaxel act synergistically to inhibit both in vitro and in vivo tumor growth of murine-derived PCC. This synergism is proposed to be through the inhibition of microtubule assembly at distinct areas, increasing the mitotic blockade of G2/M and thus, promoting cell death
[68]. Niclosamide, another anthelminthic drug, has been shown to potently decrease expression of the androgen receptor variant AR-V7, which drives castration resistant PCa (CRPC). Consequently, it has potential to sensitize treatment to enzalutamide, an anti-androgen of the second generation, which is approved for use in treatment of patients with CRPC that are no longer responsive to docetaxel
[69].
Niclosamide, an FDA-approved antihelminthic drug, exerts an anticancer effect in multiple types of cancer
[129][130]. For example, niclosamide inhibits colorectal cancer cells’ liver metastases by downregulating S100A4 and blocking the Wnt/β-catenin signaling pathway
[131][132]. It decreases lung cancer invasion by inhibiting the S100A4/NF-B/MMP9 axis: 350 reduces both the breast cancer cell pulmonary metastases and metastatic potential of lapatinib-resistant breast cancer cells by inhibiting the STAT3-FAK-Src axis
[133] and epithelial-mesenchymal transition (EMT)
[134], respectively. Furthermore, it inhibits the IL6-STAT3-AR signaling pathways in enzalutamide-resistant advanced PCa cells, reversing drug resistance and cellular invasion
[135].
3.2.7. Anti-Microbial Drugs
Minocycline, a semi-synthetic tetracycline derivative, was initially given FDA-approval for acne and some STDs, but it was later shown to have other non-antibiotic beneficial properties, including usage in inflammatory diseases
[136]. Lokeshwar showed that chemically modified tetracyclines, particularly CMT-3, was able to promote apoptosis in PCa cell lines via activation of caspase-3 and caspase-9
[72].
Antifungal itraconazole, which has a high safety profile, has been noted for its anti-angiogenesis properties
[137][138]. It inhibits mTOR, which reduces angiogenesis through the cholesterol trafficking route
[139]. Itraconazole binds to the sterol-sensing domain of NPC1 and targets the mitochondrial protein VDAC1 to regulate AMPK and MTOR, resulting in the suppression of cell proliferation and angiogenesis
[140]. In addition, it downregulates the PDGF/PI3K/Akt/mTOR pathway in infantile hemangioma
[141]. A non-comparative, randomized, phase II study was conducted to evaluate the antitumor efficacy of two doses of oral itraconazole in men with metastatic PCa. Results showed that high-dose itraconazole (600 mg/day) has a modest antitumor activity in men with metastatic CRPC
[137].
3.2.8. Anti-Malarial Drugs
Artemisinin (ARS) is a well-known antimalarial medication that is used to treat about 600 million cases of malaria every year
[142]. According to recent research, ARS and its derivatives exhibit anticancer activity as they promote non-apoptotic programmed cell death
[143][144][145]. For example, artesunate (ART)-based drugs increased ROS production and favored lysosomal over autophagic ferritin degradation in cancer cells
[146].
Similarly, chloroquine (CQ) and its derivative, hydroxychloroquine (HCQ), are antimalarial drugs that also treat several diseases, including rheumatoid arthritis, discoid lupus erythematosus, and systemic lupus erythematosus
[147][148]. CQ or HCQ, the only FDA-approved autophagy flux inhibitors, have been used to treat pancreatic and other cancers
[149][150].
Quinacrine, an antimalarial drug discovered in the 1920s, aids in the treatment of a variety of malignancies, owing to its ability to activate the p53 gene
[151]. Mainly, quinacrine induces p53 expression by facilitating the chromatin transcription (FACT) protein complex, which is trapped onto the chromatin, thereby inhibiting CK2-mediated phosphorylation of p53
[152][153].
3.2.9. Others
Zoledronic acid (ZA), a member of the bisphosphonates class of drugs, inhibits bone resorption via inhibition of osteoclast activity, hence its role in treatment of osteoporosis
[154]. This repurposed drug has been approved for clinical use in treating PCa. Corey et al. demonstrated the anti-PCa effects of ZA, both in vivo by decreasing metastatic potential and in vitro by inducing G1 arrest, proliferation decline, and apoptosis
[73]. In regard to PCa, ZA inhibits osteoblastic and osteolytic metastasis
[73], while it clinically reduces skeletal complications in PCa patients with bone metastases
[155]. Although the treatment with celecoxib, a COX-2 inhibitor, in combination with ZA therapy, is considered complementary for the treatment of advanced or metastatic PCa, it did not improve the survival rate
[156]. In contrast, the combination of ZA and docetaxel enhanced PCa patients’ survival
[128]. Based on early clinical trials, ZA is typically given every three weeks; however, current evidence suggests that giving ZA every 12 weeks results in similar outcomes in men with CRPC bone metastases
[157].
Valproic acid (VPA) is used in the treatment of epileptic, bipolar, and schizophrenic disorders by inhibiting class I histone deacetylases (HDAC)
[158]. Xia et al. demonstrated that chronic VPA administration results in reduced PCa cellular proliferation and upregulated caspase-2 and caspase-3 activity
[74].
Mifepristone, an anti-progesterone with potent progesterone receptor affinity, is used for pregnancy termination. Etreby et al. showed mifepristone’s anti-cancer effects in both androgen sensitive and insensitive PCa, via induction of TFG-β-1, a pro-apoptotic transcription factor
[75]. In fact, mifepristone’s anticancer activity is known for its ability to inhibit tumor growth via blocking the overexpressed cell surface receptors, such as progesterone, estrogen, and glucocorticoid receptors, in CRPC cells
[159]. With combined estrogen treatment, anti-progesterone delivery to PCa cells suppressed the development of androgen-insensitive PCa in vivo
[160]. The anti-tumor activity of mifepristone was reported against multiple cancer types, including androgen-sensitive and androgen-insensitive PCa
[161]. In addition, a phase II study, performed by Taplin et al., suggested that the combination therapy of mifepristone with corticosteroids, ketoconazole, or 5-α reductase inhibitors might prevent the compensatory surge in adrenal androgens in patients with CRPC
[162].
Disulfiram was first employed in the rubber vulcanization process, but it has been utilized as an alcohol-aversion medication to treat alcoholism
[163][164]. Disulfiram’s mechanisms of action are closely linked to the acetaldehyde dehydrogenase (ALDH)-related cellular metabolic processes. For example, disulfiram-mediated acetaldehyde metabolism is a viable therapeutic target for BRCA1/2-deficient cancer cells
[165]. ALDH positive atypical teratoid/rhabdoid tumor cells also had their metabolism decreased by disulfiram. Furthermore, because of its ability to block ALDH, disulfiram inhibits formaldehyde oxidation in cancer cells, resulting in cell death
[166][167]. In addition, ALDH maintains the stemness of cancer cells, which explains disulfiram’s anticancer action in PCSCs
[168][169]. Disulfiram serves as a DNA methyltransferase (DNMT1) inhibitor that restores tumor suppressor genes and DNA demethylation in PCa cells. Mainly, it reduces 5-methyl cytosine (5meC) content and methylation in APC and RARB gene promoters
[170]. Hence, in a pilot trial for recurrent PCa, Schweizer and colleagues tested for disulfiram in epigenetic therapy via evaluating 5meC content in peripheral blood mononuclear cells (PBMC)
[171]. Additionally, enhanced reduction in tumor growth was obtained upon the combination of disulfiram with Cu
2+ in CRPC xenografts
[172]. In addition, diocarb (disulfiram metabolite) and copper complex may potentially target NPL4 protein to regulate protein turnover of tumorigenesis, promoting stress-response pathways
[173].
Rapamycin has been repurposed for cancer therapy after being authorized as an immunosuppressant for kidney transplantation
[174] and an anti-restenosis drug
[175][176]. Rapamycin inhibits mTORC1 by binding to the FRB domain of mTOR and hence its use in cancer treatment
[177]. Treatment with rapamycin reduced leukemic progenitor cells in patients with acute myeloid leukemia
[178][179]. It also demonstrated efficient antitumor activity in patients with drug-resistant chronic myelogenous leukemia, with very minor adverse effects in the majority of cases
[180][181]. A study demonstrated that Rapamycin is effective in the in vitro treatment of glioblastoma and neuroblastoma by targeting their CSC population
[182].
Thalidomide, a glutamic acid derivative, was used as a sedative in 1957 and to treat morning sickness in pregnant women
[183]. Nowadays, thalidomide is widely recognized as an antiangiogenic agent that inhibits VEGF, bFGF, tumor necrosis factor-alpha (TNF-α), and various other pro-angiogenic factors
[184].
Interestingly, a study assessed the in vitro and in vivo antitumor properties and mechanisms of action of ST1926 synthetic retinoids in targeting the cancer stem-like cells population of human PCa. Results revealed that ST1926 substantially reduced proliferation of PCa cells and induced cell cycle arrest, p53-independent apoptosis, and early DNA damage. It also significantly reduced prostate spheres’ formation ability in vitro, denoting sufficient eradication of the self-renewal ability of the highly androgen-resistant CSCs
[185].