Review Article
Print
Review Article
Investigation of the promising repurposed, computational, and anticancer candidates to co-deliver with cabazitaxel for hormonal-resistance prostate cancer
expand article infoMusa Albatsh
‡ Middle East University, Amman, Jordan
Open Access

Abstract

The novel antimicrotubular taxane called cabazitaxel was developed to avoid the primary mechanism of resistance for docetaxel, the first-line treatment for patients with hormone-resistant prostate cancer (HRPC). However, its ability to increase the survival rate for HRPC is limited to a few months, and cabazitaxel-resistant prostate cancer was also developed. Consequently, this review aims to assess the repurposing, computational, and anticancer candidates that may be used in conjunction with cabazitaxel to improve its antiproliferative efficacy against prostate cancer. This work reveals that there are no additional benefits when combining cabazitaxel with chemotherapies with similar mechanisms of action, such as docetaxel. Diethylstilbestrol and ketoconazole were not recommended for use with cabazitaxel since they had modest anticancer effects and caused lethal toxicity. Computational studies have demonstrated that HRPC gene inhibitors may have promising anticancer activity against prostate cancer. Additionally, loading these combinations in transferrin-targeted nano-sized vesicles can significantly enhance their anticancer efficacy.

Keywords

cabazitaxel, combination, computational candidates, natural anticancer agents, prostate cancer, repurposed drugs

Introduction

In developed countries, prostate cancer ranks as the second leading cause of cancer-related death in the male population. Androgen deprivation therapy (ADT), such as leuprolide, goserelin, triptorelin, abiraterone, enzalutamide, and histrelin, has been indicated as the gold standard therapy for patients with early-stage, low-risk cancer as well as advanced or metastatic disease, according to the NCCN guideline (Siegel et al. 2024). Advanced prostate cancer can progress to hormone-resistant prostate cancer (HRPC) at the last stages of this cancer. Docetaxel is a main part of the treatment for patients with HRPC. However, taxane (docetaxel) resistance reduces its effectiveness; hence, cabazitaxel is indicated as an alternative treatment (Siegel et al. 2024). The purpose of the second-generation taxane, Jevtana® (cabazitaxel), is to overcome docetaxel-resistant prostate cancer. This drug has less affinity for the P-glycoprotein efflux pump, which is the main cause of docetaxel resistance. However, cabazitaxel did not show any further advantage over docetaxel in patients with HRPC (Sartor et al. 2016). Moreover, cabazitaxel (20 mg/m2), cabazitaxel (25 mg/m2), and docetaxel (75 mg/m2) were administered every three weeks to 1168 patients with HRPC along with daily prednisone. After receiving treatment with cabazitaxel and docetaxel, the median overall survival for patients with HRPC was reported to be 24.5 months for cabazitaxel at a dose of 20 mg/m2, 25.2 months for 25 mg/m2 of cabazitaxel, and 24.3 months for docetaxel at a concentration of 75 mg/m2. The incidences of developing severe adverse effects (such as febrile neutropenia, diarrhea, and hematuria) among these patients were found to be 41.2%, 60.1%, and 46.0% for 20 mg/m2 cabazitaxel, 25 mg/m2 cabazitaxel, and 75 mg/m2 docetaxel, respectively. Additionally, the toxicity profiles of docetaxel and cabazitaxel were varied; for example, cabazitaxel at a dose of 20 mg/m2 often exhibited the lowest level of toxicity (Sartor et al. 2016). The main suggested mechanisms of cabazitaxel (CBZ) resistance may include alteration in epithelial–mesenchymal transition markers, activation of extracellular signal-regulated kinase (ERK) signals (responsible for microtubule movement, tumor mitosis, and division of cancer cells) as a therapeutic target for CBZ-resistant HRPC, up-regulation of the oxytocin receptor–signaling pathway (highly expressed in DU145 resistance prostate cancer cells compared to parent DU145 cells), oncogenic stimulation of the phosphatidylinositol-3-kinase (PI3K), protein kinase B (PKB/AKT), and mammalian target of rapamycin (mTOR) pathway promoting tumor formation (Hango et al. 2018; Hongo et al. 2024). Several strategies were used to remediate resistance to chemotherapeutic agents in the treatment of advanced prostate cancer. Among these approaches, the use of drugs in combination can possibly enhance the efficacy and safety of anticancer drugs in various ways. This method can improve the efficacy of chemotherapies by reducing drug resistance through promoting the anti-proliferative efficacy of drugs with different mechanisms of action, extending the duration of action of both drugs, and reducing pharmacokinetic variability. Additionally, it can enhance the safety profile of cancer therapy by lowering the number of doses and thereby minimizing the side effects of cancer therapy. Besides, the goal of drug repurposing is to investigate the new uses of approved or understudied drugs beyond their original medical indications. This approach can shorten the time and lower the expense of creating new treatments. The cost of developing repurposed treatments is $300 million, while it costs $2–3 billion for new drugs (Bubley and Xu 2019). Therefore, this review retrospectively suggests a collection of repurposed, computational, and anticancer options to synergize the anticancer activity of CBZ against prostate cancer.

Methods

It is worth noting that the many possible repurposed medicines in the previous reviews were mainly investigated in silico or pre-clinical prostate cancer models without any human evidence. Alternatively, this review focuses on drugs for which there is support from human data. Furthermore, trials evaluating clinical cancer outcomes will be prioritized over trials evaluating biomarkers alone. This work categorizes the combination of CBZ with numerous candidates into three groups: 1) the other chemotherapies, 2) repurposed candidates, and 3) computational options.

Results and discussion

Combination of CBZ with the other chemotherapies

This section summarizes and discusses the past studies that include the use of CBZ in combination with other chemotherapy agents such as docetaxel, carboplatin, genistein, radiotherapy, antiandrogen therapy, mitoxantrone, and tasquinimod.

The previous investigations have demonstrated that the combination of dual androgen receptor-targeting medications (abiraterone + enzalutamide) with a combination of taxanes (docetaxel + CBZ) did not increase the survival rates of patients with prostate cancer. This was caused by the fact that all of these medications block the androgen receptor signaling by inhibiting the same transport mechanism, androgen receptor nuclear transport. Moreover, it was shown that the activity of docetaxel after abiraterone treatment was less than anticipated, with a median overall survival of just 12.5 months, as opposed to 19 months in the TAX327 trial (which treated patients with a combination of taxanes only). In comparison to participants in the TAX327 (48%) and a similar cohort of abiraterone-naive patients (54%), PSA responses were found to be lower (26%) (Mezynski et al. 2012; van Soest et al. 2013).

The median progression-free survival for patients with HRPC who took the CBZ and carboplatin (n = 81), an anti-neoplastic drug that suppresses DNA repair and synthesis in cancer cells, in combination (a CBZ dose of 25 mg/m2 and carboplatin of AUC 4 mg/mL per min) was enhanced from 4.5 months to 7.3 months after 31 months of following up compared with patients treated with CBZ alone ((n = 79) and a CBZ dose of 25 mg/m2). However, the grade 3–5 adverse effects, such as fatigue, anemia, neutropenia, and thrombocytopenia, were more noticeable in the combination group compared with CBZ alone, but no treatment-related deaths had occurred in both groups. Moreover, the percentages for these treatment-related adverse events were found as follows: fatigue (7 [9%] of 79 in the CBZ group vs. 16 [20%] of 81 in the combination group), anemia (3 [4%] vs. 19 [23%]), neutropenia (3 [4%] vs. 13 [16%]), and thrombocytopenia (1 [1%] vs. 11 [14%]) (Corn et al. 2019).

The IC50 of CBZ against PC3-Luc prostate cancer cells was also improved when it was used with genistein, an isoflavone compound and angiogenesis inhibitor derived from a plant called Genista tinctoria, by 2-fold with a combination index of 0.693 (where a combination index < 1 refers to synergism and > 1 confirms the antagonism) at a concentration of 5 µg/mL for genistein and 20 nM for CBZ. The in vivo results using the xenograft model of PC3-Luc cells also showed that the average tumor sizes were found to be 2008.7 ± 214.1 mm3 (control), 1913.5 ± 248.2 mm3 (genistein), 1650.8 ± 731.4 mm3 (CBZ), and 419.8 ± 249.2 mm3 (combination) (Zhang et al. 2013).

Lin et al. (2020) have also reported that the 5-year biochemical disease-free survival for 20 HRPC patients (who received triple therapies: CBZ 4 mg/m2 weekly + radiotherapy with a total dose to the prostate of 75.6 gray + second-generation antiandrogen therapy (gonadotropin-releasing hormone), namely, Enzard® (enzalutamide) for a total of 24 to 28 months with a median follow-up time of 56 months) was 73%. In contrast, four of the patients were unable to finish chemotherapy because of dose-limiting toxicities (e.g., rectal bleeding, diarrhea, and raised transaminase), and 75% of these patients had very high-risk prostate cancer (three patients increased their PSA levels and 13 patients had undetected PSA levels).

Mitoxantrone, a type II topoisomerase inhibitor, was also used to synergize the anticancer activity of CBZ against prostate cancer. Furthermore, CBZ 25 mg/m2 + Mitoxantrone 10 mg/m2 were administered on day 1 of a 21-day cycle by 23 patients with HRPC. The PSA levels were declined by 50% among 12 patients. The median duration of response was found to range between 4.9 and 10.0 months. However, the majority of these patients developed adverse events related to this combination as follows: n = 6 (sepsis and febrile neutropenia), n = 9 neutropenia, and n = 3 thrombocytopenia (Aggarwal et al. 2017).

Consistent with the above studies, PSA levels were also dropped by more than 30% in 16 out of 25 patients with HRPC who received oral tasquinimod, an immunomodulating and anti-antiangiogenic oral agent with anti-prostate cancer, at one of three escalating dose levels (0.25 mg, 0.5 mg, and 1.0 mg once daily) with 25 mg/m2 CBZ. For these patients, the median composite progression-free survival was 8.5 months. The combination-related toxicities among 15 patients include grade 3 fatigue, sensory neuropathy, atrial fibrillation, liver function, and grade 4 febrile neutropenia abnormalities (Armstrong et al. 2017). To summarize, the combination of CBZ with different chemotherapies resulted in a higher CBZ activity, but it also raised treatment-related toxicities. In patients with advanced prostate cancer, nanocarriers were crucial to deliver CBZ selectively to prostate cancer and decrease adverse events.

Combination of CBZ with repurposed drug candidates

The antiproliferative activities of ketoconazole, diethylstilbestrol, corticosteroids, digoxin, itraconazole, megestrol, disulfiram, mifepristone, statin, and metformin against prostate cancer were previously evaluated in the literature (Table 1).

Table 1.

Suggested repurposed candidates to synergize the antiproliferative efficacy of cabazitaxel (Xu and Bubley 2019; Albatsh 2022).

Drug Original Use Mechanism of Action Cancer Target Published Human Data
Celecoxib Selective Cox-2 inhibitor Inhibition of nuclear Factor κB and Akt signaling, blockage of ErbB3 Androgen receptor expression Clinical trial II/III (Smith et al. 2006)
Clinical trial II (Flamiatos et al. 2017)
Clinical trial III (Mason et al. 2017)
Retrospective data (Etheridge et al. 2018)
Dexamethasone Anti-inflammatory Reduction in the synthesis of adrenal androgen and pituitary adrenocorticotropic hormone Androgen receptor signaling Clinical trial II (Venkitaraman et al. 2015)
Diethylstilbestrol Estrogen Medical castration by negative feedback, competitive androgen receptor binding, direct cytotoxicity Androgen deprivation, having cytotoxic activity in HRPC Clinical trial II (Manikandan et al. 2005)
Clinical trial III (Shamash et al. 2011)
Digoxin Antiarrhythmic Apoptosis stimulation by intracellular calcium influx, suppression of DNA topoisomerase II, increment of interleukin 8, reduction of hypoxia-inducible factor 1α, activation of Src kinase Multiple proposed targets Retrospective data (Platz et al. 2011)
Retrospective data (Kaapu et al. 2015, 2016)
Clinical trial II (Lin et al. 2014)
Retrospective data (Zhao et al. 2021)
Disulfiram Alcohol abuse Suppression of DNMT1 and stimulation of metallothionein Triggering of APC and RARß, release of reactive oxygen species Clinical trial II (Schweizer et al. 2013)
Itraconazole Antifungal Angiogenesis deactivation, Hedgehog signaling inhibition, weak antiandrogen effect compared with ketoconazole Hedgehog pathway, blood vessel formation Clinical trial II (Antonarakis et al. 2013)
Ketoconazole Antifungal Decreasing the synthesis of androgen Blockage of CYP17A1, inhibition of androgen synthesis Retrospective data (Nakabayashi et al. 2006)
Clinical trial II (Taplin et al. 2009)
Clinical trial II (Gil-Bazo et al. 2013)
Clinical trial II (Amato et al. 2013)
Clinical trial II (Lo et al. 2015)
Clinical trial II (Millikan et al. 2003)
Clinical trial II (Ryan et al. 2007)
Clinical trial II (Mostaghel et al. 2014)
Clinical trial III, randomized, open labeled (Small et al. 2004)
Clinical trial III, randomized, open labeled (Millikan et al. 2008)
Megestrol Appetite stimulant, contraceptive Progestin Unknown Clinical trial III (Dawson et al. 2000)
Metformin Type 2 diabetes, glucose control Activation of AMPK with downstream inhibition of the mTOR signaling pathway, reduction of hepatic gluconeogenesis and systemic insulin levels Disruption of the mTOR pathway, cell proliferation apoptosis Retrospective data (Margel et al. 2013)
Clinical trial II (Rothermundt et al. 2014)
Retrospective data (Joshua et al. 2022)
Mifepristone Pregnancy termination Suppression of progesterone, androgen receptors, and dihydrotestosterone Androgen receptor blockage Clinical trial II (Hayes et al. 2006)
Statins Hypercholesterolemia Modification of androgen precursors and steroid signaling, disruption of the mevalonate pathway, and anti-inflammatory properties Unknown, but likely inhibition of androgen precursor synthesis Retrospective data (Platz et al. 2006)
Retrospective data (Yu et al. 2014)
Retrospective data (Larsen et al. 2017)
Retrospective data (Raval et al. 2016)
Clinical trial II, double-blinded (Murtola et al. 2018)
Retrospective data (Cao et al. 2023)
Retrospective data (Papagiannakis et al. 2025)

Ketoconazole, an antifungal drug, should be avoided by those who have HRPC, since it has been demonstrated to be a less potent androgen synthesis inhibitor and has been associated with side effects such as weakness, edema, and diarrhea (Millikan et al. 2003, 2008; Small et al. 2004; Nakabayashi et al. 2006; Ryan et al. 2007; Taplin et al. 2009; Amato et al. 2013; Gil-Bazo et al. 2013; Mostaghel et al. 2014; Lo et al. 2015).

Diethylstilbestrol, an estrogen hormone, has been indicated as an alternative option for orchiectomy in patients with hormone-dependent prostate cancer. Because it works through several mechanisms, including competitive inhibition of the androgen receptor and direct anticancer efficacy in prostate cells, it can also be utilized to treat individuals with HRPC. However, it can result in lethal cardiac events (Manikandan et al. 2005; Shamash et al. 2011).

Corticosteroids such as dexamethasone are given as part of a supportive care therapy plan for patients with advanced prostate cancer who have pain, weakness, and anorexia. Furthermore, they have weak anticancer effects in prostate cancer by decreasing the synthesis of testosterone (Venkitaraman et al. 2015).

Digoxin, a widespread antiarrhythmic drug, has recently suppressed the growth of prostate cancer by 25% through triggering many apoptotic processes, including enhanced interleukin 8, hypoxia-inducible factor 1 inhibition, intracellular calcium influx, and Src kinase activation (Platz et al. 2011; Lin et al. 2014, 2020; Kaapu et al. 2015, 2016). Although digoxin has been linked to a decreased incidence of prostate cancer, retrospective cohort studies have found that digoxin did not correlate to prostate cancer mortality (HR 1.17; 95% CI 0.88–1.57) (Flahavan et al. 2014). Furthermore, there were ten clinical studies conducted on 108,444 participants (15,835 of them were digoxin users). Six studies found a substantial decrease in prostate cancer risk associated with digoxin use (adjusted RR = 0.892, 95% CI: 0.799–0.997, p = 0.044) compared to nonusers. While four of them have revealed a significant correlation between digoxin and greater prostate cancer-specific mortality compared to controls (adjusted HR = 1.142, 95% CI: 1.005–1.297). There was no statistical heterogeneity observed in this study (all I2 < 50%, p > 0.1) (Zhao et al. 2021). In conclusion, digoxin had a preventive action on the risk of prostate cancer in men. However, digoxin use was linked to a higher risk of prostate cancer-related mortality. More well-designed studies are needed to adequately interpret the causation of these findings.

Clinical trial studies have demonstrated that itraconazole, megestrol, disulfiram, and mifepristone can be effective in treating prostate cancer. These medications exert their anticancer efficacy through several mechanisms, including blocking the androgen receptor, decreasing the release of reactive oxygen species, and inhibiting the formation of new blood vessels (Dawson et al. 2000; Hayes et al. 2006; Antonarakis et al. 2013; Schweizer et al. 2013).

In 2006, the first epidemiological investigation indicated that statin users had a 61% lower incidence of deadly prostate cancer (RR 0.39; 95% CI 0.19–0.77) than nonusers. The largest study to date had around 11,772 men with nonmetastatic prostate cancer with 1791 deaths during follow-up. After controlling for potential confounders, post-diagnostic clinical follow-up had shown that statin was associated with a 24% decreased risk of cancer-specific death (hazard ratio, HR 0.76; 95% CI 0.66–0.88) (Stopsack et al. 2016; Cao et al. 2023; Papagiannakis et al. 2025). A recent study found that males who took statins had a longer time to progress on ADT compared to nonusers (Dos Santos et al. 2024).

According to in vitro studies, metformin can suppress prostate and other cancers through several mechanisms, involving effects on both cancerous and non-cancerous cells. Two high-quality population-based studies evaluated the relationships between metformin use and death following a diagnosis of prostate cancer. In Ontario, Canada, Margel et al. (2013) first examined 3837 diabetic males who were over 66 years old and had prostate cancer. A 24% decreased risk of prostate cancer-specific death was linked to every extra six months of metformin use (95% CI 11–36%). Lower all-cause mortality was also substantially linked to metformin. Bensimon et al. (2014) found no evidence of overall prostate cancer-specific mortality (RR 1.09; 95% CI 0.51–2.33) in their trial, which involved 935 males from the United Kingdom. A recent meta-analysis of all studies that took diabetes diagnosis into account found that metformin was linked to better overall survival and lower biochemical recurrence among patients with prostate cancer (Joshua et al. 2022; de Oliveira et al. 2025). However, the results for mortality specific to prostate cancer were inconsistent, but the study of Margel et al. (2013) was the most dominant.

Combination of CBZ with promising, naturally derived anticancer agents

As usual, plants provide an abundant supply of the taxane chemotherapeutics, including CBZ. This section explores the wide spectrum of natural compounds, such as isoflavones, naphthoquinones, and polyphenols, for which anticancer action against prostate cancer has been identified. Furthermore, these agents can exert their anticancer activity via various mechanisms such as inhibition of angiogenesis, reducing the growth and metastasis of prostate cancer cells, targeting tumor suppressor genes, decreasing the production of androgenic hormones, and using photodynamic properties of some plant families.

Previous investigations have shown that the protocatechuic acid from Punica granatum, polysaccharides from mushrooms Ganoderma lucidum, Trametes versicolor, Grifola frondosa, and Rubus occidentalis, as well as resveratrol from Vitis vinifera, mainly induced apoptosis and impaired the metastasis of prostate cancer cells by inhibition of the angiogenesis of prostate tumor cells (Panda et al. 2022). Besides, the additional mechanisms for Punica granatum (pomegranate) were also involved in reducing oxidative stress biomarkers in tissue, stimulation of apoptosis executor caspase 3, accumulation of reactive oxygen species, and inhibition of matrix metalloproteinases (MMP2/MMP9) that control the migration and invasion of cancer cells (Deng et al. 2017; Johari et al. 2022). The IC50 of docetaxel and flutamide (nonsteroidal antiandrogen used primarily to treat prostate cancer) against PC3-Luc and LNCaP prostate cancer cells was reduced by 50% following a combination with Ganoderma lucidum (Rahimnia et al. 2023). The latest study has shown that Ganoderma lucidum can also target signal transducer and activator of transcription 3 (STAT3), which is involved in the proliferation and metastasis of prostate tumor cells (Zhou et al. 2024). The immune system (mainly T-cells and splenic natural killer cells) could also be targeted using Trametes versicolor and Grifola frondosa mushrooms as adjuvants with docetaxel (Habtemariam 2020; He et al. 2022; Li et al. 2024; Lamkhade et al. 2025). Perez (2020) and Fuloria et al. (2022) revealed that Vitis vinifera had not only inhibited angiogenesis but also arrested the G1 phase cell cycle and promoted the activity of apoptosis-associated enzymes. Prostate cancer cell growth was not inhibited by Rubus occidentalis (black raspberry extract), cyanidin-3-rutinoside, or protocatechuic acid during the in vitro study (Eskra et al. 2019).

In this study, it has also investigated whether isoflavones such as genistein and daidzein as well as methoxylated ones (formononetin, biochanin A, and glycitein), which are less polar than compounds containing hydroxyl groups, may be used in conjunction with CBZ to treat hormone-dependent prostate cancer. The anticancer efficacy of genistein is linked to its ability to block angiogenesis. In the meantime, other isoflavones (such as daidzein) have been shown in publications to have antiangiogenic properties. Genistein and daidzein have been suggested to affect the expression of genes that guarantee angiogenesis and metastasis, such as matrix metalloproteinases (ΜΜP-2, ΜΜP-9, ММ-11, МΜ-13, ΜΜ-14), EGF, ANGPT2, and CTGF (Rabiau et al. 2010). Interestingly, Batra et al. (2020) have also found that the testosterone levels of LNCaP and PC-346C CaP prostate cell lines were reduced by approximately 3-fold. Similarly, it has been found that genistein can also interfere with the therapeutic effects of the estrogen receptor antagonist anticancer agent tamoxifen in athymic nude mice (Du et al. 2012). Besides, formononetin was also one of the isoflavones that suppressed proliferation, migration, and invasion of prostate cancer cells. This anticancer effect may occur by inducing expression of a tumor suppressor gene known as early growth response protein 1 (EGR1) and raising the levels of the cell adhesion molecule E-cadherin protein (Liang et al. 2022). In accordance, the percentages of cell viability for PC3-Luc cells were reduced in a time-dose-dependent way after these cells were treated (after 48 hours) with 40 µM of formononetin from 80% to 30% (Wang et al. 2020). Biochanin-A has also demonstrated inhibitory action in prostate cancer by (1) blocking tyrosine kinase events in the signal transduction pathway and (2) disrupting the metabolism of testosterone via stimulating the activity of the enzyme uridine 5-diphospho-glucuronosyltransferase (UDPGT) (Anuranjana et al. 2023; Park et al. 2024). Unfortunately, no research has yet clarified the primary anticancer mechanism of glycitein against prostate cancer.

Undoubtedly, naphthoquinones (like plumbagin, juglone, and droserone) along with anthraquinones such as the hypericin family have anticancer properties. Furthermore, the cellular uptake of plumbagin, a naphthoquinone with anticancer properties obtained from the plant leadwort, loaded within transferrin-bearing liposomes by B16-F10 mouse melanoma, A431 human epidermoid carcinoma, and T98G human brain tumor cells was improved by at least 1.4-fold compared with control liposomes and 2-fold compared with the drug solution. In addition, the intravenous injection of transferrin-bearing liposomes containing plumbagin led to 10% tumor suppression and an additional 10% tumor regression when compared to untargeted liposomes or untreated groups, respectively (Sakpakdeejaroen et al. 2019). In accordance with plumbagin, the survival rates for PC3-Luc and DU145 cells were reduced (after 72 hours) following treatment with 500 μg/mL of juglone up to 46% and 53%, respectively (Mahdavi et al. 2019). No prior study evaluated the potential anticancer activity of droserone against prostate cancer. Among anthraquinones, hypericin and other dianthrones obviously attract attention, especially when used in chemotherapy simultaneously with radiation based on the photodynamic characteristics of hypericins (hypericin, pseudohypericin, protohypericin). For example, the IC50 of the hexane flower extract of Hypericum perforatum L. against PC3-Luc and LNCaP prostate cancer cells was reduced after 72 hours of treatment from 47.04 ± 4.63 µg/mL to 11.06 ± 0.87 µg/mL (for PC3-Luc cells) and from 58.24 ± 0.03 µg/mL to 11.70 ± 0.80 µg/mL (for DU145 cells) (Petrović et al. 2022).

The encapsulation of epigallocatechin-3-gallate, a green tea polyphenol with anti-tumor effects, within transferrin-conjugated vesicles also led to a marked increase in the cellular uptake of this drug (by at least 1.5-fold) in all three cell lines (A431 cells, B16-F10 melanoma cells, T98G fibroblast cancer cells) compared to that reported in control vesicles and the drug solution. Besides, the in vitro anti-proliferative efficacy on cells treated with epigallocatechin-3-gallate loaded in transferrin-bearing vesicles was significantly enhanced when compared to control vesicles, by 1.9-fold for A431 cells, 2.7-fold for B16-F10 cells, and 4-fold for T98G cancer cells (with IC50 ranging from 0.36 ± 0.05 to 1.41 ± 0.17 g/mL for the transferrin-bearing vesicles). Tumor suppression was also observed in 40% of A431 and B16-F10 cancer cells upon intravenous administration of transferrin-bearing vesicles containing epigallocatechin-3-gallate. In addition, animal survival was increased by more than 20 days in comparison to controls (Lemarié et al. 2013).

Combination of CBZ with computational candidates

Computational methods have also been used to investigate potential drug repositioning options for the treatment of HRPC. Kim et al. (2019) used Gene Expression Omnibus, a database repository of high-throughput gene expression data, to acquire gene expression data from cancerous and normal prostate tissue samples from HRPC patients. Moreover, a meta-signature analysis using the metaDE R-project® program, statistical computing software, was used to identify the expressed genes in HRPC. This software simulates and generates the outcomes based on existing data and research methodologies. For instance, it was shown that the treatment options sorafenib, olaparib, elesclomol, tanespimycin, and ponatinib were successful in decreasing the expression of genes related to HRPC, such as MYL9, E2F2, APOE, and ZFP36, with an IC50 of less than 10 µM for all of drugs. Vascular endothelial growth factor (VEGF) plasma levels were found to be significantly greater in HRPC patients than in localized disease patients (Kim et al. 2019). VEGF is also attributed to the progression of other types of cancer. Pazopanib is a multi-targeted VEGF tyrosine kinase inhibitor. It was discovered that pazopanib and lenalidomide have synergistic cytotoxicity in multiple myeloma (Podar et al. 2006). Kim et al. (2019) found that a combination of lenalidomide and pazopanib had the highest synergistic effect with a combination index value less than 0.6 (where a combination index less than 1 indicates the synergism) (Table 2).

Table 2.

The promising computational drug candidates for cabazitaxel (Kim et al. 2019; Albatsh 2022).

Drug name Mechanism of action in HRBC Original indication Activity in HRPC
Elesclomol Targeting (APOE) HRBC gene and induction for the oxidative stress by increasing the synthesis of reactive oxygen species within prostate cancer cells. Adjuvant treatment with paclitaxel in patients with metastatic melanoma. Active (IC50 < 10 μM)
Emetin Suppression of protein biosynthesis, DNA interaction, and regulation of pro-apoptotic factors Amoebiasis (a protozoan infection) and emetic drug. Active (IC50 < 10 μM)
Fluoxuridine Inhibition of thymidylate synthetase production, blockage of the synthesis of thymidylic acid, and reduction of DNA synthesis, as well as binding of 5-flurodeoxyuridine triphosphate into DNA and formation of single-strand breaks. Gastrointestinal adenocarcinomas, breast, and ovarian cancers. Active (IC50 < 10 μM)
Geldanamycin Deactivation of hypoxia-inducible factor 1 and heat shock protein 90 involved in cellular responses to hypoxia, glycolysis, cycle, growth, survival, apoptosis, and angiogenesis of prostate cancer cells. Chronic myeloid leukemia. Active (IC50 < 10 μM)
Olaparib Targeting (E2F2) HRPC gene and inhibition of enzyme polyribose polymerase. Breast and ovarian cancers. Active (IC50 < 10 μM)
Ponatinib Targeting ZFP36 HRPC gene. Chronic myeloid leukemia and acute lymphoblastic leukemia. Active (IC50 < 10 μM)
Sorafenib Blockage of VEGF and rapidly accelerated fibrosarcoma (RAF) kinases and targeting (MYL9) HRPC gene. Advanced renal cell carcinoma, hepatocellular carcinomas, and thyroid cancer. Active (IC50 < 10 μM)
Tanespimycin Suppression of the production of Hsp90, a stabilizer for proteins required for prostate tumor growth. Chronic myeloid leukemia and solid tumors. Active (IC50 < 10 μM)
Trichostatin A Inhibition of epidermal growth factor receptor pathway mediated in regulation of cell growth, differentiation, motility, adhesion, and tumorigenesis. Fungal infections Active (IC50 < 10 μM)
Lenalidomide + Pazopanib Immunomodulation (lenalidomide) and suppression of tyrosine kinase (pazopanib) for prostate cancer cells. Lenalidomide (Multiple myeloma) + Pazopanib (Metastatic renal cell carcinoma and advanced soft tissue sarcomas). Combination index < 0.6

Discussion and evidence-based medicine

Clinicians often do not rely only on observational studies for introducing new medications to patients due to ambiguities in risk predictions and residual confounding. Additional questions beyond the scope of this review should be addressed in clinical practice. The majority of studies only provide predictions on the association between drug use and cancer-specific mortality. There is typically a lack of information on which patient subgroups will benefit the most. Certain prescription drugs may cause malignant transformation, metastatic formation, or castration resistance. Drugs may target specific molecular abnormalities, resulting in significant benefits for a subset of patients. Predictive biomarkers for such reactions should be identified and used in future trials. As a result, clinically significant variations may exist based on cancer biology, stage, grade, main treatments, and comorbidities. The timing of treatment initiation, the optimal dosage, and the side effects for prostate cancer patients have not been adequately assessed for any of the examined drugs. Polypharmacy and drug interactions are an essential concern. They may impact cancer outcomes beyond mortality, such as cancer incidence or biochemical progression-free survival. In polypharmacy, channeling bias might also alter pharmacoepidemiologic risk estimates for a specific drug. Besides, co-medication can affect the extent of drugs specifically directed at prostate cancer. Hence, further research should include well-designed observational studies to determine which drugs have a clinically significant impact on specific subgroups of prostate cancer patients. Population-based studies, rather than hospital-based cohorts, provide simpler and more generalizable answers to methodologically challenging questions. Eventually, suitably powered RCTs in subsets of patients may be needed.

Conclusion

The first and second generations of taxanes—docetaxel and cabazitaxel—used for treating HRPC, have developed resistance through various mechanisms. To address this issue, this review strongly suggests using CBZ in combination with several agents that have different mechanisms of action, such as other chemotherapies, repurposed drugs, and computational drugs. This strategy can bypass and compensate for the resistance mechanisms of CBZ, sustain its half-life, control the variation in pharmacokinetics, and minimize the toxicities for this chemotherapy. In contrast, past studies have shown that combining CBZ with other anticancer drugs targeting the same transport receptors or similar sites of action did not improve survival rates in HRPC patients. Active targeting of nano-sized vesicles entrapping repurposed drugs with transferrin ligand enhanced the in vitro anti-proliferative efficacy of these drugs and resulted in in vivo tumor regression. Computational methods are promising and may help identify an effective synergistic combination with CBZ. To gain the maximum benefit from a combination strategy, further in vitro and in vivo studies, as well as human clinical trials, are needed to develop effective and safe combinations of CBZ. Moreover, tumor-targeted nanocarriers are vital for selectively delivering these combinations to prostate cancer cells with minimal toxicity.

Additional information

Conflict of interest

The author has declared that no competing interests exist.

Ethical statements

The author declared that no clinical trials were used in the present study.

The author declared that no experiments on humans or human tissues were performed for the present study.

The author declared that no informed consent was obtained from the humans, donors or donors’ representatives participating in the study.

The author declared that no experiments on animals were performed for the present study.

The author declared that no commercially available immorta­lised human and animal cell lines were used in the present study.

Funding

No funding was reported.

Author contributions

The author solely contributed to this work.

Author ORCIDs

Musa Albatsh https://orcid.org/0009-0007-9411-4912

Data availability

All of the data that support the findings of this study are available in the main text.

References

  • Aggarwal R, Bryce A, Ryan CJ, Harzstark A, Derleth C, Kim W, Friedlander T, Lin AM, Rodvelt-Bagchi T, Dhawan M, Zhang L (2017) A multicenter phase I study of cabazitaxel, mitoxantrone, and prednisone for chemotherapy-naïve patients with metastatic castration-resistant prostate cancer: A department of defense prostate cancer clinical trials consortium study. Urologic Oncology: Seminars and Original Investigations 4: 149–157. https://doi.org/10.1016/j.urolonc.2016.11.005
  • Albatsh M (2022) Development of novel tumor-targeted nanomedicines for the treatment of docetaxel-resistant prostate cancer. PhD Thesis, University of Strathclyde, Glasgow, United Kingdom. https://stax.strath.ac.uk
  • Amato RJ, Saxena S, Stepankiw M (2013) Phase II trial assessing granulocyte-macrophage—colony stimulating factor, ketoconazole plus mitoxantrone in metastatic castration-resistant prostate cancer progressing after docetaxel treatments. Cancer Investigation 3: 177–182. https://doi.org/10.3109/07357907.2013.764564
  • Antonarakis ES, Heath EI, Smith DC, Rathkopf D, Blackford AL, Danila DC, King S, Frost A, Ajiboye AS, Zhao M, Mendonca J (2013) Repurposing itraconazole as a treatment for advanced prostate cancer: a noncomparative randomized phase II trial in men with metastatic castration-resistant prostate cancer. The Oncologist 2: 163–173. https://doi.org/10.1634/theoncologist.2012-314
  • Anuranjana PV, Beegum F, Divya KP, George KT, Viswanatha GL, Nayak PG, Kanwal A, Kishore A, Shenoy RR, Nandakumar K (2023) Mechanisms behind the pharmacological application of Biochanin-A: a review. F1000Research 12: 107–148. https://doi.org/10.12688/f1000research.126059.1
  • Armstrong AJ, Humeniuk MS, Healy P, Szmulewitz R, Winters C, Kephart J, Harrison MR, Martinez E, Mundy K, Halabi S, George D (2017) Phase Ib trial of cabazitaxel and tasquinimod in men with heavily pretreated metastatic castration resistant prostate cancer (mCRPC): the CATCH trial. The Prostate 4: 385–395. https://doi.org/10.1002/pros.23277
  • Batra N, Sam A, Woldemariam T, Talbott G, de Vere White RW, Ghosh PM, Gaikwad NW, Kotchoni SO, Vinall RL (2020) Genistein combined polysaccharide (GCP) can inhibit intracrine androgen synthesis in prostate cancer cells. Biomedicines 8: 282–298. https://doi.org/10.3390/biomedicines8080282
  • Bensimon L, Yin H, Suissa S, Pollak MN, Azoulay L (2014) The use of metformin in patients with prostate cancer and the risk of death. Cancer Epidemiology, Biomarkers & Prevention 10: 2111–2118. https://doi.org/10.1158/1055-9965.EPI-14-0056
  • Cao Z, Yao J, He Y, Lou D, Huang J, Zhang Y, Chen M, Zhou Z, Zhou X (2023) Association between statin exposure and incidence and prognosis of prostate cancer: a meta-analysis based on observational studies. American Journal of Clinical Oncology 7: 323–334. https://doi.org/10.1097/COC.0000000000001012
  • Corn PG, Heath EI, Zurita A, Ramesh N, Xiao L, Sei E, Li-Ning-Tapia E, Tu SM, Subudhi SK, Wang J, Wang X (2019) Cabazitaxel plus carboplatin for the treatment of men with metastatic castration-resistant prostate cancers: a randomized, open-label, phase 1–2 trial. The Lancet Oncology 10: 1432–1443. https://doi.org/10.1016/S1470-2045(19)30408-5
  • de Oliveira RK, Prieto GB, da Cruz Silva V, Bavaresco PS, Alves NP, de Queiroz DA, de Queiroz EA (2025) Anti-tumor effect of metformin in oncological patients: a systematic review. Scientific Electronic Archives 1: 1–13. https://doi.org/10.36560/18120252025
  • Deng Y, Li Y, Yang F, Zeng A, Yang S, Luo Y, Zhang Y, Xie Y, Ye T, Xia Y, Yin W (2017) The extract from Punica granatum (pomegranate) peel induces apoptosis and impairs metastasis in prostate cancer cells. Biomedicine & Pharmacotherapy 93: 976–984. https://doi.org/10.1016/j.biopha.2017.07.008
  • Dos Santos DZ, Elbaz M, Branchard E, Schormann W, Brown CE, Meek AR, Njar VC, Hamilton RJ, Reed MA, Andrews DW, Penn LZ (2024) Sterol-like drugs potentiate statin-triggered prostate cancer cell death by inhibiting SREBP2 nuclear translocation. Biomedicine & Pharmacotherapy 177: 116934–116947. https://doi.org/10.1016/j.biopha.2024.116934
  • Eskra JN, Dodge A, Schlicht MJ, Bosland MC (2020) Effects of black raspberries and their constituents on rat prostate carcinogenesis and human prostate cancer cell growth in vitro. Nutrition and Cancer 4: 672–685. https://doi.org/10.1080/01635581.2019.1650943
  • Etheridge T, Liou J, Downs TM, Abel EJ, Richards KA, Jarrard DF (2018) The impact of celecoxib on outcomes in advanced prostate cancer patients undergoing androgen deprivation therapy. American Journal of Clinical and Experimental Urology 3: 123–132. https://e-century.us/files/ajceu/6/3/ajceu0074385.pdf
  • Flahavan EM, Sharp L, Bennett K, Barron TI (2014) A cohort study of digoxin exposure and mortality in men with prostate cancer. British Journal of Urology International 2: 1–11. https://doi.org/10.1111/bju.12287
  • Flamiatos JF, Beer TM, Graff JN, Eilers KM, Tian W, Sekhon HS, Garzotto M (2017) Cyclooxygenase-2 (COX-2) inhibition for prostate cancer chemoprevention: double-blind randomised study of pre-prostatectomy celecoxib or placebo. British Journal of Urology 5: 709–716. https://doi.org/10.1111/bju.13612
  • Fuloria S, Sekar M, Khattulanuar FS, Gan SH, Rani NN, Ravi S, Subramaniyan V, Jeyabalan S, Begum MY, Chidambaram K, Sathasivam KV (2022) Chemistry, biosynthesis and pharmacology of viniferin: potential resveratrol-derived molecules for new drug discovery, development and therapy. Molecules 16: 5072–5105. https://doi.org/10.3390/molecules27165072
  • Gil-Bazo I, Arévalo E, Castillo A, Zudaire ME, Carranza OE, Fusco JP, Castañón E, Collado-Gómez V, López I, Gil-Aldea I (2013) Safety and efficacy of maintenance therapy with a nonspecific cytochrome P17 inhibitor (CYP17i) after response/stabilization to docetaxel in metastatic castration-resistant prostate cancer. Clinical Genitourinary Cancer 2: 78–84. https://doi.org/10.1016/j.clgc.2012.11.003
  • Hayes JH, Oh WK, Kantoff PW, Manola JB, Smith MR, Gelmann EP, Bubley G, Balk SP, Taplin M (2006) Mifepristone (RU-486) in androgen independent prostate cancer. Journal of Clinical Oncology 18: 14508–14508. https://doi.org/10.1200/jco.2006.24.18_suppl.14508
  • Hongo H, Kosaka T, Oya M (2018) Analysis of cabazitaxel-resistant mechanism in human castration-resistant prostate cancer. Cancer Science 9: 2937–2945. https://doi.org/10.1111/cas.13729
  • Hongo H, Kosaka T, Takayama KI, Baba Y, Yasumizu Y, Ueda K, Suzuki Y, Inoue S, Beltran H, Oya M (2024) G-protein signaling of oxytocin receptor as a potential target for cabazitaxel-resistant prostate cancer. PNAS Nexus 3: 1–12. https://doi.org/10.1093/pnasnexus/pgae002
  • Joshua AM, Armstrong A, Crumbaker M, Scher HI, de Bono J, Tombal B, Hussain M, Sternberg CN, Gillessen S, Carles J, Fizazi K (2022) Statin and metformin use and outcomes in patients with castration-resistant prostate cancer treated with enzalutamide: A meta-analysis of AFFIRM, PREVAIL and PROSPER. European journal of Cancer 170: 285–295. https://doi.org/10.1016/j.ejca.2022.04.005
  • Kaapu KJ, Ahti J, Tammela TL, Auvinen A, Murtola TJ (2015) Sotalol, but not digoxin is associated with decreased prostate cancer risk: a population-based case–control study. International Journal of Cancer 5: 1187–1195. https://doi.org/10.1002/ijc.29470
  • Kaapu KJ, Murtola TJ, Talala K, Taari K, Tammela TL, Auvinen A (2016) Digoxin and prostate cancer survival in the Finnish Randomized Study of Screening for Prostate Cancer. British Journal of Cancer 11: 1289–1295. https://doi.org/10.1038/bjc.2016.328
  • Lamkhade SV, Pathan NH, Lad R, Jadhav NA (2024) Mushroom derived bioactive compound as anticancer agent. World Journal of Pharmaceutical Research 1: 1510–1530.
  • Larsen SB, Dehlendorff C, Skriver C, Dalton SO, Jespersen CG, Borre M, Brasso K, Nørgaard M, Johansen C, Sørensen HT, Hallas J (2017) Postdiagnosis statin use and mortality in Danish patients with prostate cancer. Journal of Clinical Oncology 29: 3290–3297. https://doi.org/10.1200/JCO.2016.71.8981
  • Lemarié F, Chang CW, Blatchford DR, Amor R, Norris G, Tetley L, McConnell G, Dufès C (2013) Antitumor activity of the tea polyphenol epigallocatechin-3-gallate encapsulated in targeted vesicles after intravenous administration. Nanomedicine 2: 181–192. https://doi.org/10.2217/nnm.12.83
  • Li X, Ruan Q, Yang W, Tian H, Wu N, Qadir J, Wang J, Hu H, Liu Y, Cai M, Yang BB (2024) Polysaccharide isolated from Grifola frondosa eliminates myeloid-derived suppressor cells and inhibits tumor growth by enhancing T cells responses. International Journal of Biological Sciences 2: 664–670. https://doi.org/10.7150/ijbs.85276
  • Lin J, Zhan T, Duffy D, Hoffman-Censits J, Kilpatrick D, Trabulsi EJ, Lallas CD, Chervoneva I, Limentani K, Kennedy B, Kessler S (2014) A pilot phase II Study of digoxin in patients with recurrent prostate cancer as evident by a rising PSA. American Journal of Cancer Therapy and Pharmacology 1: 21–32. https://pmc.ncbi.nlm.nih.gov/articles/PMC4287984/pdf/nihms634526.pdf
  • Lin J, Den RB, Greenspan J, Showalter TN, Hoffman-Censits JH, Lallas CD, Trabulsi EJ, Gomella LG, Hurwitz MD, Leiby B, Dicker AP (2020) Phase I trial of weekly cabazitaxel with concurrent intensity modulated radiation and androgen deprivation therapy for the treatment of high-risk prostate cancer. International Journal of Radiation Oncology* Biology* Physics 5: 939–947. https://doi.org/10.1016/j.ijrobp.2019.11.418
  • Lin TP, Fan YH, Chen YC, Huang WJ (2020) Digoxin lowers the incidence of prostate cancer: A nationwide population-based study. Journal of the Chinese Medical Association 4: 377–381. https://doi.org/10.1097/JCMA.0000000000000288
  • Lo EN, Beckett LA, Pan CX, Robles D, Suga JM, Sands JM, Lara PN (2015) Prospective evaluation of low-dose ketoconazole plus hydrocortisone in docetaxel pre-treated castration-resistant prostate cancer patients. Prostate Cancer and Prostatic Diseases 2: 144–148. https://doi.org/10.1038/pcan.2015.2
  • Mahdavi M, Azadbakht M, Vahdati A, Shokrzadeh M, Farhadi A (2019) Cytotoxic effects of juglone and Pterocarya fraxinifolia on prostate cancer cells. Journal of Pharmacy and Bioallied Sciences 3: 195–204. https://doi.org/10.4103/jpbs.JPBS_203_18
  • Manikandan R, Srirangam SJ, Pearson E, Brown SC, O’Reilly P, Collins GN (2005) Diethylstilboestrol versus bicalutamide in hormone refractory prostate carcinoma: a prospective randomized trial. Urologia Internationalis 3: 217–221. https://doi.org/10.1159/000087797
  • Margel D, Urbach DR, Lipscombe LL, Bell CM, Kulkarni G, Austin PC, Fleshner N (2013) Metformin use and all-cause and prostate cancer–specific mortality among men with diabetes. Journal of Clinical Oncology 25: 3069–3075. https://doi.org/10.1200/JCO.2012.46.7043
  • Mason MD, Clarke NW, James ND, Dearnaley DP, Spears MR, Ritchie AW, Attard G, Cross W, Jones RJ, Parker CC, Russell JM (2017) Adding celecoxib with or without zoledronic acid for hormone-naïve prostate cancer: long-term survival results from an adaptive, multiarm, multistage, platform, randomized controlled trial. Journal of Clinical Oncology 14: 1530–1541. https://doi.org/10.1200/JCO.2016.69.0677
  • Mezynski J, Pezaro C, Bianchini D, Zivi A, Sandhu S, Thompson E, Hunt J, Sheridan E, Baikady B, Sarvadikar A, Maier G (2012) Antitumor activity of docetaxel following treatment with the CYP17A1 inhibitor abiraterone: clinical evidence for cross-resistance? Annals of Oncology 11: 2943–2947. https://doi.org/10.1093/annonc/mds119
  • Millikan R, Thall PF, Lee SJ, Jones D, Cannon MW, Kuebler JP, Wade III J, Logothetis CJ (2003) Randomized, multicenter, phase II trial of two multicomponent regimens in androgen-independent prostate cancer. Journal of Clinical Oncology 5: 878–883. https://doi.org/10.1200/JCO.2003.04.057
  • Millikan RE, Wen S, Pagliaro LC, Brown MA, Moomey B, Do KA, Logothetis CJ (2008) Phase III trial of androgen ablation with or without three cycles of systemic chemotherapy for advanced prostate cancer. Journal of Clinical Oncology 36: 5936–5942. https://doi.org/10.1200/JCO.2007.15.9830
  • Mostaghel EA, Nelson PS, Lange P, Lin DW, Taplin ME, Balk S, Ellis W, Kantoff P, Marck B, Tamae D, Matsumoto AM (2014) Targeted androgen pathway suppression in localized prostate cancer: a pilot study. Journal of Clinical Oncology 3: 229–237. https://doi.org/10.1200/JCO.2012.48.6431
  • Murtola TJ, Syvälä H, Tolonen T, Helminen M, Riikonen J, Koskimäki J, Pakarainen T, Kaipia A, Isotalo T, Kujala P, Tammela TL (2018) Atorvastatin versus placebo for prostate cancer before radical prostatectomy—a randomized, double-blind, placebo-controlled clinical trial. European Urology 6: 697–701. https://doi.org/10.1016/j.eururo.2018.06.037
  • Nakabayashi M, Xie W, Regan MM, Jackman DM, Kantoff PW, Oh WK (2006) Response to low-dose ketoconazole and subsequent dose escalation to high-dose ketoconazole in patients with androgen-independent prostate cancer. Cancer: Interdisciplinary International Journal of the American Cancer Society 5: 975–981. https://doi.org/10.1002/cncr.22085
  • Panda SK, Sahoo G, Swain SS, Luyten W (2022) Anticancer activities of mushrooms: a neglected source for drug discovery. Pharmaceuticals 2: 176–201. https://doi.org/10.3390/ph15020176
  • Papagiannakis A, Kwiatkowski M, Wyler SF, Mortezavi A, Manka L, Wettstein MS, Grobholz R, Hammerer-Lercher A, Eberli D, Prause LW (2025) Influence of Regular Statin Intake on Prostate-Specific Antigen Values, Prostate Cancer Incidence and Overall Survival in a Prospective Screening Trial (ERSPC Aarau). Cancer Medicine 1: 70485–70494. https://doi.org/10.1002/cam4.70485
  • Park Y, Jo G, Hyun H (2024) A Tumor Targeting Strategy of Phytoflavonoid Biochanin A for Efficient Fluorescence-Guided Chemotherapy. Small Science 8: 2400111–2400121. https://doi.org/10.1002/smsc.202400111
  • Petrović N, Ergün S, Đorđić-Crnogorac M, Stanojković T, Mališić E, Matić Ivana Z (2022) Hypericum perforatum L. extracts exert cytotoxic effects and show different miRNA signatures in PC-3 and DU 145 prostate cancer cells. Genetika 3: 1249–1270. http://dx.doi.org/10.2298/GENSR2203249P
  • Podar K, Tonon G, Sattler M, Tai YT, LeGouill S, Yasui H, Ishitsuka K, Kumar S, Kumar R, Pandite LN, Hideshima T (2006) The small-molecule VEGF receptor inhibitor pazopanib (GW786034B) targets both tumor and endothelial cells in multiple myeloma. Proceedings of the National Academy of Sciences 51: 19478–19483. https://doi.org/10.1073/pnas.0609329103
  • Platz EA, Leitzmann MF, Visvanathan K, Rimm EB, Stampfer MJ, Willett WC, Giovannucci E (2006) Statin drugs and risk of advanced prostate cancer. Journal of the National Cancer Institute 24: 1819–1825. https://doi.org/10.1093/jnci/djj499
  • Platz EA, Yegnasubramanian S, Liu JO, Chong CR, Shim JS, Kenfield SA, Stampfer MJ, Willett WC, Giovannucci E, Nelson WG (2011) A novel two-stage, transdisciplinary study identifies digoxin as a possible drug for prostate cancer treatment. Cancer Discover 1: 68–77. https://doi.org/10.1158/2159-8274.CD-10-0020
  • Rahimnia R, Akbari MR, Yasseri AF, Taheri D, Mirzaei A, Ghajar HA, Farashah PD, Baghdadabad LZ, Aghamir SM (2023) The effect of Ganoderma lucidum polysaccharide extract on sensitizing prostate cancer cells to flutamide and docetaxel: an in vitro study. Scientific Reports 1: 18940–18954. https://doi.org/10.1038/s41598-023-46118-8
  • Rabiau N, Kossaï M, Braud M, Chalabi N, Satih S, Bignon YJ, Bernard-Gallon DJ (2010) Genistein and daidzein act on a panel of genes implicated in cell cycle and angiogenesis by polymerase chain reaction arrays in human prostate cancer cell lines. Cancer Epidemiology 2: 200–206. https://doi.org/10.1016/j.canep.2009.12.018
  • Raval AD, Thakker D, Negi H, Vyas A, Salkini MW (2016) Association between statins and clinical outcomes among men with prostate cancer: a systematic review and meta-analysis. Prostate Cancer and Prostatic Diseases 2: 151–162. https://doi.org/10.1038/pcan.2015.58
  • Rothermundt C, Hayoz S, Templeton AJ, Winterhalder R, Strebel RT, Bärtschi D, Pollak M, Lui L, Endt K, Schiess R, Rüschoff JH (2014) Metformin in chemotherapy-naive castration-resistant prostate cancer: a multicenter phase 2 trial (SAKK 08/09). European Urology 3: 468–474. https://doi.org/10.1016/j.eururo.2013.12.057
  • Ryan CJ, Weinberg V, Rosenberg J, Fong L, Lin A, Kim J, Small EJ (2007) Phase II study of ketoconazole plus granulocyte-macrophage colony-stimulating factor for prostate cancer: effect of extent of disease on outcome. The Journal of Urology 6: 2372–2377. https://doi.org/10.1016/j.juro.2007.08.011
  • Sakpakdeejaroen I, Somani S, Laskar P, Mullin M, Dufès C (2019) Transferrin-bearing liposomes entrapping plumbagin for targeted cancer therapy. Journal of Interdisciplinary. Nanomedicine 2: 54–71. https://doi.org/10.1002/jin2.56
  • Schweizer MT, Lin J, Blackford A, Bardia A, King S, Armstrong AJ, Rudek MA, Yegnasubramanian S, Carducci MA (2013) Pharmacodynamic study of disulfiram in men with non-metastatic recurrent prostate cancer. Prostate Cancer and Prostatic Diseases 4: 357–361. https://doi.org/10.1038/pcan.2013.28
  • Shamash J, Powles T, Sarker SJ, Protheroe A, Mithal N, Mills R, Beard R, Wilson P, Tranter N, O’brien N, McFaul S (2011) A multi-centre randomised phase III trial of Dexamethasone vs Dexamethasone and diethylstilbestrol in castration-resistant prostate cancer: immediate vs deferred Diethylstilbestrol. British Journal of Cancer 4: 620–628. https://doi.org/10.1038/bjc.2011.7
  • Small EJ, Halabi S, Dawson NA, Stadler WM, Rini BI, Picus J, Gable P, Torti FM, Kaplan E, Vogelzang NJ (2004) Antiandrogen withdrawal alone or in combination with ketoconazole in androgen-independent prostate cancer patients: a phase III trial (CALGB 9583). Journal of Clinical Oncology 6: 1025–1033. https://doi.org/10.1200/JCO.2004.06.037
  • Smith MR, Manola J, Kaufman DS, Oh WK, Bubley GJ, Kantoff PW (2006) Celecoxib versus placebo for men with prostate cancer and a rising serum prostate-specific antigen after radical prostatectomy and/or radiation therapy. Journal of Clinical Oncology 18: 2723–2728. https://doi.org/10.1200/JCO.2005.03.7804
  • Taplin ME, Regan MM, Ko YJ, Bubley GJ, Duggan SE, Werner L, Beer TM, Ryan CW, Mathew P, Tu SM, Denmeade SR (2009) Phase II study of androgen synthesis inhibition with ketoconazole, hydrocortisone, and dutasteride in asymptomatic castration-resistant prostate cancer. Clinical Cancer Research 22: 7099–7105. https://doi.org/10.1158/1078-0432.CCR-09-1722
  • van Soest RJ, van Royen ME, de Morrée ES, Moll JM, Teubel W, Wiemer EA, Mathijssen RH, de Wit R, van Weerden WM (2013) Cross-resistance between taxanes and new hormonal agents abiraterone and enzalutamide may affect drug sequence choices in metastatic castration-resistant prostate cancer. European Journal of Cancer 18: 3821–3830. https://doi.org/10.1016/j.ejca.2013.09.026
  • Venkitaraman R, Lorente D, Murthy V, Thomas K, Parker L, Ahiabor R, Dearnaley D, Huddart R, De Bono J, Parker C (2015) A randomized phase 2 trial of dexamethasone versus prednisolone in castration-resistant prostate cancer. European Urology 4: 673–679. https://doi.org/10.1016/j.eururo.2014.10.004
  • Wang R, Li K, Xie Z, Wang B, Dai Y, Chen J, Ye Y (2020) Formononetin induces apoptosis of PC-3 human prostate cancer cells via regulating long noncoding RNA H19 and the mitochondrial apoptosis pathway. Pharmacognosy Magazine 69: 1–6.
  • Yu O, Eberg M, Benayoun S, Aprikian A, Batist G, Suissa S, Azoulay L (2014) Use of statins and the risk of death in patients with prostate cancer. Journal of Clinical Oncology 1: 5–11. https://doi.org/10.1200/JCO.2013.49.4757
  • Zhang S, Wang Y, Chen Z, Kim S, Iqbal S, Chi A, Ritenour C, Wang YA, Kucuk O, Wu D (2013) Genistein enhances the efficacy of cabazitaxel chemotherapy in metastatic castration-resistant prostate cancer cells. The Prostate 15: 1681–1689. https://doi.org/10.1002/pros.22705
  • Zhao S, Li X, Wu W, Liu S, Shen M, Zhang Z, He J (2021) Digoxin reduces the incidence of prostate cancer but increases the cancer-specific mortality: a systematic review and pooled analysis. Andrologia 11: 14217–14227. https://doi.org/10.1111/and.14217
  • Zhou X, Wang J, Guo Y, Lai H, Cheng S, Chen Z, Li H, Li Q, Mao X (2024) Water extract of sporoderm-broken spores of Ganoderma lucidum elicits dual antitumor effects by inhibiting p-STAT3/PD-L1 and promoting ferroptosis in castration-resistant prostate cancer. Journal of Functional Foods 113: 106018–106032. https://doi.org/10.1016/j.jff.2024.106018
login to comment