eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
1/2024
vol. 28
 
Share:
Share:
Review paper

Application of nitroxoline in urologic oncology – a review of evidence

Wojciech Tomczak
1
,
Wojciech Krajewski
2
,
Joanna Chorbińska
2
,
Łukasz Nowak
2
,
Jan Łaszkiewicz
1
,
Katarzyna Grunwald
1
,
Szymon Pisarski
1
,
Adam Chełmoński
1
,
Bartosz Małkiewicz
2
,
Tomasz Szydełko
1

  1. University Centre of Excellence in Urology, Wrocław Medical University, Wrocław, Poland
  2. Department of Minimally Invasive and Robotic Urology, University Centre of Excellence in Urology, Wrocław Medical University, Wrocław, Poland
Contemp Oncol (Pozn) 2024; 28 (1): 1–8
Online publish date: 2024/05/19
Article file
Get citation
 
PlumX metrics:
 

Introduction

Cancer-related mortality represents the second leading cause of premature death globally, with its incidence rising steadily – from 5.6 million cases in 1990 to 10.8 million in 2019 [1]. In the United States alone, the year 2023 is expected to see 609,820 deaths and nearly 2.0 million new cancer diagnoses [2]. This escalating issue has catalysed a surge in the exploration of novel treatment strategies. A notable approach is the concept of drug repurposing, wherein established pharmaceuticals, previously utilised in distinct medical domains, are being reassessed for their efficacy, in this case in oncology. This methodology is particularly advantageous due to the well-documented safety profiles and the established spectrum of side effects of these drugs, a critical consideration given the typically high toxicity associated with oncological therapies [3]. In this realm, the antibiotic nitroxoline (NIT), initially introduced in the 1960s, has emerged as a compound of significant interest, demonstrating potential therapeutic benefits in the treatment of cancer [4]. Currently, the potential for NIT as a treatment option is being explored across various cancers, including ones originating from the bladder, prostate, pancreas, lungs, brain, and other sites [510]. This review investigates the use of NIT in urologic cancer treatment, discusses potential adverse effects, and digs into specific mechanisms of action within the framework of urologic oncology.

Material and methods

In this systematic review, we present a comprehensive analysis of the therapeutic potential of NIT in urologic oncology, underscored by strict adherence to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines and the population, intervention, comparison, and outcome (PICO) framework. We formulated the following PICO question to guide our inquiry: In humans with cancer, animal models, and tumour cell lines (P), how does nitroxoline treatment, either alone or supplemented to existing regimens (I), compare to regimens without nitroxoline (C), in terms of the response to therapy in humans or any observed effects on tumour in animal models and cell lines (O)?

Two independent researchers conducted a systematic literature search of the PubMed, Medline, and Embase databases using the following search string: Nitroxoline AND (oncology OR cancer OR tumor OR tumour). The inclusion criteria were limited to original studies published in English language. Studies published in languages other than English, as well as reviews, conference reports, case-reports, letters to the publisher, and comments, were excluded. Additionally, a cross-reference search was carried out. The process of literature search and selection is transparently detailed in the PRISMA flow chart below (Fig. 1). The publication has been recorded in the international prospective register of systematic reviews (PROSPERO) and assigned the code CRD42024502266. To find as many reports of NIT adverse events (AEs) as possible, we performed additional searches for clinical trials unrelated to oncology.

Fig. 1

The preferred reporting items for systematic reviews and meta-analyses flow-chart presenting the entire literature search process

/f/fulltexts/WO/54116/WO-28-54116-g001_min.jpg

Safety of administration

Chemotherapy-associated adverse effects (CAAEs) resulting in a decline of quality of life are the main cause of discontinuation of effective regimens. Additionally, some CAAEs develop years after the completion of treatment. Others lead to infertility, neuropathies, secondary malignancies, and severe organ toxicities [11]. In light of the aforementioned, the development of novel treatment strategies must be balanced between efficacy and patient safety. In this context, a repurposing strategy is particularly enticing because most considered medicines have already been in use for decades. This is also the case with NIT.

The vast majority of clinical studies on NIT have been conducted in the context of urinary tract infections (UTIs). Naber et al. identified 32 clinical trials related to UTIs (4 of which were randomised and 28 were not) [12]. Of these, 29 reported on AEs. To this date, no severe AEs have been reported in various clinical settings [13].

Upon reviewing the outcomes of 4 high-quality, multicentre, single-blind randomised controlled trials that included 466 patients, Naber et al. found that 9.4% of participants undergoing NIT therapy encountered AEs, as opposed to 7.8% in the control group, although this difference was not statistically significant. Gastrointestinal AEs such as nausea, stomach pain, and diarrhoea were the most frequently reported. Headache, itching, and eyelid swelling were reported; however, these were isolated incidents. A total of 5 individuals terminated their treatment due to these AEs, with 3 from the NIT group and 2 from the control group [12].

Within the scope of 28 controlled and uncontrolled clinical trials, a total of 1354 individuals were involved. Among these, 6 trials involving 478 adult participants and 7 trials including 182 paediatric subjects revealed no complications. However, some AEs were reported in the remaining ones. Gastrointestinal complaints, particularly nausea, vomiting, and diarrhoea were the most frequently mentioned, with prevalence rates spanning 1.3–9% across trials. One study including 60 adults reported itching in 3.3% of cases. Exanthema was noted in 2.2% of participants in another study. In CTs conducted on children, nausea was the sole AE reported, affecting up to 10% of the paediatric population [12].

A solitary clinical case of NIT overdose has been identified. It holds significance, especially in the context of oncologic treatment regimens where in general higher dosages are administered. The aforementioned case involved an individual with suicidal intentions who consumed 5000 mg of NIT. Upon admission to the hospital, the patient exhibited only symptoms of fatigue while remaining alert and oriented. No further toxicity signs were noted. The patient recovered without the need for any specific medical intervention [14].

Bladder

Bladder cancer (BC) ranks as the 10th most common malignancy globally [15, 16] with data suggesting that 50–70% of on-muscle invasive bladder cancer (NMIBC) patients experience recurrence, and 30% progress to MIBC [10, 17, 18]. The 2023 European Association of Urology guidelines recommend transurethral resection of the bladder tumour followed by intravesical instillation of bacille Calmette-Guerin (BCG) therapy as the standard treatment for NMIBC. However, the effectiveness of BCG therapy is limited in some patients due to drug resistance or intolerable side effects [19]. Furthermore, some BCs already invade the muscle at diagnosis, complicating treatment strategies. Likewise, the resistance to standard chemotherapeutics like doxorubicin (DOX) and cisplatin [10]. The genomic heterogeneity of urothelial cancers is one explanation for these diverse responses and treatment failures, emphasising the need for novel drugs and their tailored utilisation. This also highlights the challenges in preventing high recurrence and addressing the progression to MIBC, which contribute significantly to incidence and mortality.

Angiogenesis

Many development and progression of human diseases rely on angiogenesis [2023]. This is also the case with cancer. Its significance in tumour growth and metastasis is well understood; therefore, inhibiting angiogenesis has emerged as a promising technique for slowing tumour growth and increasing the efficacy of cytotoxic anticancer treatments [24, 25]. Shim et al. investigated the type 2 methionine aminopeptidase (MetAP2) pathway and sirtuins as the 2 most probable mechanisms for angiogenesis inhibition. Human umbilical vein endothelial cells (HUVEC), human BC KU7 line, and human breast cancer HCC1954 were evaluated.

The type 2 methionine aminopeptidase protein has been widely identified and extensively studied as a molecular target for angiogenesis inhibitors [2629]. The type 2 methionine aminopeptidase inhibition activates p53, a tumour suppressor, and increases cyclin-dependent kinase inhibitor 1A. As a result, the cyclin E1 and cyclin-dependent kinase 2 (CDK2) complex, which phosphorylates retinoblastoma 1 (RB1) and regulates endothelial cell G1-S cell cycle transition, are inhibited [30, 31]. Moreover, increasing evidence links silent information regulators (sirtuins) to cellular ageing and angiogenesis [32]. In vivo, SIRT1 suppression causes endothelial cell senescence and prevents blood vessel development [33, 34].

Shim et al. performed 2 concurrent screening assays: one targeting MetAP2 with a 175,000-compound chemical library, and another utilising HUVEC against the Johns Hopkins Drug Library [35]. These screens were aimed at discovering new inhibitors of angiogenesis. Substances indicated by both drug libraries were designated for further testing. One of the most potent double hits was NIT. Firstly, scientists cultured HUVEC with different concentrations of NIT to verify its potency of proliferation inhibition. Secondly, to verify the mechanism of action (MoA) of NIT, small inter-interfering RNAs (siRNAs) targeting MetAP2 and SIRT1 were transfected into HUVEC. Changes in p53, p21, and RB1 protein expression were assessed. The results of the study group matched the positive control. Nitroxoline demonstrated potent and highly selective inhibition of MetAP2. Additionally, a dose-dependent inhibition of SIRT1 and SIRT2 activity was observed when treated with NIT. Conversely, NIT exerted no changes in SIRT3.

Following the experiments on cell lines, an in vivo Matrigel plug assay was performed on mice. One group received a carrier solution, the other NIT, both administered intraperitoneally once a day. After 3 days of pre-treatment, a Matrigel plug containing pro-angiogenic factors was implanted subcutaneously near the abdominal midline. Treatment continued for an additional 7 days. Subsequently, the plugs were harvested and stained. Compared with the control, NIT significantly reduced the formation of new microvessels.

Next, Human breast cancer xenograft has been explored. HCC1954 cells were injected subcutaneously. Once the tumours were noticeable, the mice were grouped based on mass size to ensure equal average volume across the 2 groups. The first group received a carrier solution, and the other was treated with NIT, both administered intraperitoneally every other day. Mass volume was monitored throughout the study. After 30 days, the mice were euthanised and tumours were collected for analyses. Treatment with NIT resulted in a 60% reduction in volume and a 43% decrease in tumour weight compared to the controls. Furthermore, NIT therapy increased the acetylation of the TP53 protein in tumour tissues, indicating an inhibition of SIRT1 activity. Furthermore, the levels of MetAP2 substrate Met-14-3-3 as well as p53, p21, and RB1 were elevated, implying that NIT suppresses this pathway activity in vivo. Immunohistochemical analysis revealed a 58% reduction in angiogenesis in the NIT-treated group.

Lastly, an orthotopic BC model was employed. KU7 cells were orthotopically injected into murine bladders. Three days later, the mice were treated orally with either NIT or placebo for 2 weeks. Multiple bioluminescence imaging showed that NIT treatment significantly reduced tumour growth compared to the placebo. The histopathological analysis further demonstrated a noticeable decrease in tumour size in the NIT group. Immunoblot analysis of bladder tissues was consistent with in vitro results. No significant difference in SIRT1-related protein levels was detected between groups, and acetylated TP53 (acetyl-K382) levels were undetectable in both groups, precluding further analysis of TP53. These differences certainly highlight the cell line-specific MoA of NIT and emphasise the necessity for additional investigation.

Drug resistance

Drug resistance is a significant challenge in cancer treatment, with about 80–90% of cancer-related deaths being directly or indirectly linked to it [36]. Additionally, nearly all drugs used in oncology have encountered some form of resistance, undermining the effectiveness of chemotherapy regimens [36]. One of the most frequently described mechanisms of resistance is drug efflux from the site of action. It has been described in most types of cancer and it is believed that the ATP-binding cassette transporter protein family is responsible for it [3740].

Lin et al. explored this mechanism of resistance through the prism of BC, identifying overexpression of P-glycoprotein (P-gp) as the root cause [10]. Encoded by the MDR1 gene, it reduces intracellular concentrations of its substrates, such as DOX, by actively transporting them out of tumour cells, leading to decreased drug efficacy [41].

Evidence suggests that inhibiting P-gp may also increase CIS sensitivity. This implicates P-gp involvement in reducing tumour cell sensitivity to non-P-gp substrates (like CIS) [42]. Lin et al. also suggested that STAT3 influences MDR1 gene expression [10]. Additionally, a growing body of research describes an association between the overexpression STAT3 and BC poor prognosis. Therefore, STAT3 inhibition has been suggested to effectively downregulate P-gp expression [43, 44].

In his study, Lin et al. conducted both in vitro and in vivo investigations demonstrating the reversal of P-gp overexpression mediated by NIT application [10]. Scientists utilised human BC cell line T24, alongside DOX-resistant (T24/DOX) and CIS-resistant (T24/CIS). The T24 and T24/DOX cells were exposed to increasing concentrations of DOX, whereas T24 and T24/CIS cells were treated with CIS to evaluate the impact of NIT on cancer proliferation. This involved treating all cell lines with growing concentrations of NIT. The analysis encompassed drug sensitivity, cell cycle distribution, apoptosis examination, and western blot analysis. In animal xenograft models, tumour growth and body weight were monitored, followed by the harvesting of tumours for histological analysis.

A dose- and time-dependent reduction in viability was observed in the T24 cells and the drug-resistant lines upon treatment with NIT. Notably, as the NITs concentration increased, a higher proportion of cells remained in the G0/G1 phase across the T24, T24/DOX, and T24/CIS cells. Additionally, the percentage of cells undergoing apoptosis increased with the NIT concentration, reaching up to 62% in the drug-resistant lines. Western blot analysis revealed a dose-dependent suppression of STAT3 signalling and P-gp expression.

In the following section of the study, a subcutaneous xenograft model was employed in mice utilising T24/DOX and T24/CIS cells. The experimental groups received an oral course of NIT therapy at a dosage that the authors declared to be derived from the standard dosage for UTIs [45]. The control received phosphate-buffered saline. Notably, NIT treatment resulted in significant tumour size reduction, with corresponding decreases in average tumour weights. Tumour growth rates were slower in the NIT groups, with no significant changes in the body weight of the mice, indicating low regimen toxicity. Histological analysis showed distinct morphological changes, including cancer cell necrosis in the NIT group. Western blotting confirmed reduced levels of phosphorylated STAT3 (the most active form), P-gp, and Mcl-1 in NIT-treated tumours, but the change in total STAT3 expression was not statistically significant.

Reversing epithelial mesenchymal transition

Metastatic BC remains a formidable challenge in clinical oncology, marking a transition to an advanced stage associated with severely reduced survival rates. Furthermore, treating metastatic BC presents a considerable challenge due to the heterogeneity of metastatic lesions. Among these, numerous “private” mutations exist, and they keep evolving individually. Therefore, understanding the underlying mechanisms of metastasis development is of utmost importance [46]. Medical fitness for palliative chemotherapy in metastatic BC is another major concern; repurposing drugs with low toxicity would raise treatability [47].

Mechanism verification

In 2020, Xu et al. carried out a study to investigate the MoA of NIT in BC [48]. They conducted in vitro cell line experiments (MBT-2 and J82) as well as an animal model with C3H/He mice. Although numerous NIT MoAs were identified, scientists attempted to verify one previously undescribed in BC, i.e. the one involving the blockade of epithelial-mesenchymal transition (EMT). The process is crucial in embryonic development, tissue fibrosis, wound healing, and notably in cancer metastasis [49, 50]. Some of the key molecules in EMT include matrix metalloproteinases (MMPs) such as MMP-2 and MMP-9, which promote cancer cell migration and extracellular matrix degradation.

During the initial stage of the study, reduced viability of BC lines was observed in both dose- and time-dependent manners. Furthermore, treatment with NIT led to noticeable apoptotic changes, such as chromatin condensation and nuclear fragmentation, as compared to controls. Next, transwell and wound-healing assays showed significant NIT efficacy in impairing cell migration.

In the following section of this study, the impact of NITs on the expression of proteins related to EMT was investigated. The analysis showed a decrease in the levels of N-cadherin, slug, and MMP-9 in both cell lines. Howe-ver, the reduction in MMP-2 expression was observed only in MBT-2 cells. Moreover, NIT treatment led to a decrease in the expression of Bcl-2, an anti-apoptotic protein, while increasing the levels of cleaved caspase-3 (C-caspase-3), a pro-apoptotic protein.

Lastly, mice with subcutaneously implanted BC cells were investigated. Two treatment arms received different NIT regimens via intragastric administration over 12 days. The higher NIT dose significantly reduced tumour volume and weight when compared with the controls. Tumour section immunohistochemistry showed that the higher-dose treatment arm led to larger necrotic areas within the tumours and decreased cell proliferation, as indicated by lower expression of Ki-67, a marker for proliferation, and CD31, an endothelial marker indicating blood vessel density. Additionally, MMP-9 expression, associated with tumour invasion and metastasis, was significantly reduced in the high-dose group compared to the controls.

Molecular pathway identification

In 2022 Ren et al. attempted to establish the mechanism accountable for the aforementioned EMT inhibition [19]. As confirmed by many non-coding RNAs, circRNAs in particular contribute to processes such as cell proliferation, apoptosis, and cancer metastasis. Their impact was evaluated on the following cell lines: T24, UM-UC-3, TCCSUP, and J82.

Consistent with the prior findings, all cell lines employed in the study exhibited decreased migration capability as well as impaired healing abilities after NIT treatment. Next, using a circRNA microarray, molecules that were co-expressed and upregulated across all treated cell lines were identified. Then, using specific primers, circNDRG1 was identified as the molecule consistently elevated across all 4 cell lines, thereby prompting its selection for further exploration.

Building on this, they created 2 unique siRNAs that specifically target the circNDRG1. This small inter-interfering RNAs mediated reduction of circNDRG1 was achieved in 2 types of BC cells: T24 and UM-UC-3. Such altered cells have been evaluated using transwell and wound-healing assays. Reducing circNDRG1 increased cell spreading abilities. To further validate this, a plasmid increasing the expression of circNDRG1 was introduced into the T24 and UM-UC-3 cells. The cells’ metastatic ability decreased, indicating the protective function of circNDRG1.

Lastly, the efficacy of NIT in mitigating cancer metastasis was evaluated using a murine model. After introducing UM-UC-3 cells, marked with a luminescent molecule, into mice via tail vein injection, the subjects were treated with NIT. The IVIS imaging system showed increased luminescence intensity in mice with reduced circNDRG1 levels, indicating a greater cancer spread than in the control group.

To completely understand the pathway behind circNDRG1 expression. Ren et al. examined whether it may sponge BC-associated miRNA. Starbase, CircInteractome, and CircBank prediction tools were used to anticipate which miRNA circNDRG1 would target. miR-520h was the only microRNA predicted by all 3 tools. Further, the transwell and wound-healing assay showed that miR-520h increased the migration of BC cells, suggesting that circNDRG1’s MoA is associated with miR-520h. The next step was to identify miR-520h’s target gene. Using predictions from four databases (miRmap, starBase, TargetScan, and miRDB), a list of 172 potential genes was established. Smad7, known to be involved in EMT, was selected. Subsequent investigations demonstrated that treating cells with NIT increased smad7 levels and inhibited EMT. Ultimately, reducing smad7 expression in BC cells counteracted the anti-metastatic effects of the miR-520h inhibition, demonstrating that miR-520h facilitates cancer spread by targeting smad7 and modulating the EMT pathway.

Prostate

Prostate cancer (PCa) holds the position of the second most diagnosed cancer worldwide [51]. Notwithstanding, from 2007 to 2014, there was a sharp decline in PCa incidence, largely due to changes in screening guidelines that led to reduced testing for prostate-specific antigen (PSA) and subsequent overdiagnosis reduction. However, since 2014, there has been an approximate annual increase of 3% in the overall incidence rates, with advanced-stage PCa incidence rates climbing by 5% each year [52]. Despite overdiagnosis and overtreatment being a major concern, metastatic PCa still presents as largely incurable and resistant to the latest regimens. Around 6% of new PCa cases are metastatic at diagnosis and frequently androgen independent [53, 54]. The magnitude of this issue is underlined by the fact that there has been no significant progress in increasing the survival rates for men with advanced PCa in the past 20 years [55]; as a result, NIT is now part of the repurposing inventory for PCa treatment.

Cell cycle arrest and apoptosis induction

The 2015 study by Chang et al. delves into the potential of NIT repurposing as a novel treatment against both hormone-sensitive and hormone-refractory PCa cells targeting LNCaP, PC-3, and DU-145 cell lines [9]. The research focuses on how NIT impacts the AMP-activated protein kinase (AMPK)/mTOR signalling pathway and interacts with Chk2 activation in the fight against PCa.

AMP-activated protein kinase acts as a crucial cellular regulator, overseeing essential functions such as glucose absorption, fatty acid oxidation, and more [56, 57]. Its role in halting cancer cell growth by changing energy balance makes it a target for cancer therapy. Alongside Chk2, serine/threonine protein, a kinase activated in response to DNA damage, plays a fundamental role in the cellular DNA damage response [58, 59], either facilitating DNA repair or enforcing cell cycle arrest and triggering apoptosis, preventing the spread of genetically compromised cells [60, 61].

The researchers discovered that NIT triggers G1 arrest of the cell cycle and subsequent death of PCa cells via an AMPK-dependent mechanism [9]. Nitroxoline was demonstrated to block both the mTOR-p70S6K signalling pathway and the cyclin D1-Rb-Cdc25A axis. Furthermore, NIT triggered AMPK-dependent activation of Chk2, which was followed by a shift from pro-survival DNA repair activity to pro-apoptotic one. This at least partially contributes to cancer cell death. Overall, NIT inhibited cell growth in hormone-sensitive and hormone-refractory cell lines in a time-dependent manner. Furthermore, NIT-treated cell lines demonstrated increased autophagy rates, as shown by a rise in the well-known autophagy-related protein LC3-II [62]. This shows that NIT may play a role in the development of therapies for PCa.

To further exclude possible confounding effects on PCa cells exerted by NIT’s chelating abilities, additional investigations have been conducted. Iron and zinc – cofactors in hundreds of processes which regulate the cell cycle – might be influenced by NIT’s chelating ability. This is particularly useful in eliminating bacterial infections [63]. A theory assuming the influence of chelating properties on the viability of PCa cell lines was also verified. The anticancer action of NIT was found to be unaffected by ZnCl2 supplementation. As a result, NITs MoA in PCa cells were shown to be independent of its ability to chelate ions.

PD-1 blockade and nitroxoline synergy

In the 2019 research conducted by Xu et al., the efficacy of combining NIT with PD-1 blockade therapy in the treatment of PCa was explored [64]. This investigation involved in vitro studies using human and murine PCa cell lines, as well as in vivo studies within orthotopic animal models. The cell lines used were human PCa lines LNCaP, DU145, and PC3, and the normal epithelial prostate cell line RWPE-1. Additionally, the study incorporated the murine PCa cell line RM9. The RM9 cells were genetically modified to express luciferase and human PSA, creating the RM9-Luc-PSA cells. This particular cell line is advantageous for dynamically tracking drug responses and tumour progression.

PD-1 receptor, known as programmed death 1, is a receptor located on the surface of T-cells, playing a crucial role in the immune system’s checkpoint mechanisms [65]. It functions to prevent autoimmune reactions by controlling the intensity of immune responses, thus minimising potential tissue damage [66]. Programmed death-ligand 1 (PD-L1) is a ligand that binds to the PD-1 receptor. It is found on the surface of various cell types, including some cancer cells. The interaction between PD-1 and PD-L1 leads to the suppression of T-cell activity, allowing tumours with high PD-L1 expression to evade the immune system’s attack [67]. This evasion facilitates the growth and spread of the tumour by shielding it from immune-mediated destruction. Concluding, PD-L1 expression is associated with a poor prognosis, adverse outcomes, and higher recurrence rates [6871].

In PCa murine models, Xu et al. examined the effectiveness of treatments using NIT, PD-1 blockade, and a combination of both (NIT + PD-1 blockade). Through in vivo imaging, it was found that the effectiveness of NIT matched that of PD-1 blockade when compared to a control group. Additionally, reductions in serum PSA levels were observed in each treatment arm, with the most significant decreases seen in the NIT + PD-1 blockade. This treatment also resulted in the lowest tumour size and weight. Furthermore, examination of histological sections from resected tumours revealed that microvascular density was notably reduced in the group receiving the dual-drug treatment. The findings suggest that combining NIT and PD-1 blockade enhances the anti-tumour response across all evaluated treatment effects.

In the in vitro analyses, NIT notably decreased the viability of PCa cell lines RM9-Luc-PSA, LNCaP, DU145, and PC3 over time. On the other hand, no significant cytotoxic effects were observed in normal RWPE-1 cells. Subsequent investigations focused on NIT’s impact on cell cycle progression and apoptosis. Nitroxoline treatment altered the expression of key cell cycle regulatory molecules, leading to cell cycle arrest in the analysed PCa lines. Specifically, NIT reduced the levels of CDK2, CDK6, and cyclin D3 in a dose-dependent manner. The absence of these molecules results in the arrest of the cell cycle [64]. Moreover, researchers discovered that NIT also lowers the levels of Bcl-2 and Bcl-xL while enhancing the expression of caspase-3 – proteins intimately linked to apoptosis. This pattern of expression supports the induction of apoptosis.

Conclusions

Drug repurposing is reshaping how new treatments are developed, significantly cutting the costs and time required to bring a new one to the market, while also leveraging the known safety profile of this medication. Quinolone derivatives, including NIT, have gained considerable interest in the field of oncology. The application of NIT in urologic oncology shows great potential. However, different cell lines have been shown to respond differently to NIT treatment. We believe that the next step in evaluating the effectiveness of novel, tailored therapies using NIT is to perform genomic profiling of numerous cell lines. This will involve identifying molecular profiles that can predict the success or failure of NITs in therapeutic application. Similar research initiatives are currently examining new pharmaceuticals and certain types of cancer [7274]. In parallel with the development of other innovative drugs, this approach could substantially enhance patient treatment outcomes through targeted therapy regimens based on detailed molecular profiling of their disease.

To provide a comprehensive overview of the role of NIT in urological oncology, it is important to acknowledge that certain studies focused on NIT derivatives were not covered by this review. Notwithstanding, these substances yielded encouraging results when used to study renal cell carcinoma [75]. Furthermore, NIT is currently being evaluated in a multicentre phase II clinical trial for NMIBC, registered under the trial identifier CTR20131716. Taking it all into consideration, we believe it is just a matter of time before NIT or one of its novel derivatives gains traction in oncology.

Disclosures

Institutional review board statement

Not applicable.

Assistance with the article

None.

Financial support and sponsorship

None.

Conflicts of interest

None.

References

1 

Roser M, Ritchie H. Cancer. Our world in data. Available from: https://ourworldindata.org/cancer (accessed: 26.032024).

2 

Common Cancer Sites – Cancer Stat Facts. Available from: https://seer.cancer.gov/statfacts/html/common.html (accessed: 26.03.2024).

3 

Zhang Q, Wang S, Yang D, Pan K, Li L, Yuan S. Preclinical pharmacodynamic evaluation of antibiotic nitroxoline for anticancer drug repurposing. Oncol Lett 2016; 11: 3265-3272.

4 

Mitrović A, Kos J. Nitroxoline: repurposing its antimicrobial to antitumor application. Acta Biochim Pol 2019; 66: 521-531.

5 

Li G, Zheng YH, Xu L, et al. BRD4 inhibitor nitroxoline enhances the sensitivity of multiple myeloma cells to bortezomib in vitro and in vivo by promoting mitochondrial pathway-mediated cell apoptosis. Ther Adv Hematol 2020; 11: 2040620720932686.

6 

Yu JG, Ji CH, Shi MH. Nitroxoline induces cell apoptosis by inducing MDM2 degradation in small-cell lung cancer. Kaohsiung J Med Sci 2019; 35: 202-208.

7 

Veschi S, Ronci M, Lanuti P, et al. Integrative proteomic and functional analyses provide novel insights into the action of the repurposed drug candidate nitroxoline in AsPC-1 cells. Sci Rep 2020; 10: 2574.

8 

Lazovic J, Guo L, Nakashima J, et al. Nitroxoline induces apoptosis and slows glioma growth in vivo. Neuro Oncol 2015; 17: 53-62.

9 

Chang WL, Hsu LC, Leu WJ, Chen CS, Guh JH. Repurposing of nitroxoline as a potential anticancer agent against human prostate cancer: a crucial role on AMPK/mTOR signaling pathway and the interplay with Chk2 activation. Oncotarget 2015; 6: 39806-39820.

10 

Lin W, Sun J, Sadahira T, et al. Discovery and validation of nitroxoline as a novel STAT3 inhibitor in drug-resistant urothelial bladder cancer. Int J Biol Sci 2021; 17: 3255-3267.

11 

Lustberg MB, Kuderer NM, Desai A, Bergerot C, Lyman GH. Mitigating long-term and delayed adverse events associated with cancer treatment: implications for survivorship. Nat Rev Clin Oncol 2023; 20: 527-542.

12 

Naber KG, Niggemann H, Stein G, Stein G. Review of the literature and individual patients’ data meta-analysis on efficacy and tolerance of nitroxoline in the treatment of uncomplicated urinary tract infections. BMC Infect Dis 2014; 14: 628.

13 

Sobke A, Makarewicz O, Baier M, et al. Empirical treatment of lower urinary tract infections in the face of spreading multidrug resistance: in vitro study on the effectiveness of nitroxoline. Int J Antimicrobial Agents 2018; 51: 213-220.

14 

MIP Pharma Polska Sp. z o.o. Charakterystyka Produktu Leczniczego: Nitroxolin forte. Available from: http://chpl.com.pl/#detail=18203174!83317814 (accessed: 28.03.2024)

15 

Ploeg M, Aben KKH, Kiemeney LA. The present and future burden of urinary bladder cancer in the world. World J Urol 2009; 27: 289-293.

16 

Bladder cancer statistics | World Cancer Research Fund International. WCRF International. Available at: https://www.wcrf.org/cancer-trends/bladder-cancer-statistics/ (accessed: 28.032024).

17 

Fernandez-Gomez J, Madero R, Solsona E, et al. Predicting nonmuscle invasive bladder cancer recurrence and progression in patients treated with bacillus Calmette-Guerin: the CUETO scoring model. J Urol 2009; 182: 2195-2203.

18 

Sylvester RJ, van der Meijden APM, Oosterlinck W, et al. Predic-ting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 2006; 49: 466-465; discussion 475-477.

19 

Ren L, Jiang M, Xue D, et al. Nitroxoline suppresses metastasis in bladder cancer via EGR1/circNDRG1/miR-520h/smad7/EMT signaling pathway. Int J Biol Sci.2022; 18: 5207-5220.

20 

Colville-Nash PR, Scott DL. Angiogenesis and rheumatoid arthritis: pathogenic and therapeutic implications. Ann Rheum Dis 1992; 51: 919-925.

21 

Ashton N. Retinal angiogenesis in the human embryo. Br Med Bull 1970; 26: 103-106.

22 

Folkman J, Merler E, Abernathy C, Williams G. Isolation of a tumor factor responsible for angiogenesis. J Exp Med 1971; 133: 275-288.

23 

Bhat S, Shim JS, Zhang F, Chong CR, Liu JO. Substituted oxines inhibit endothelial cell proliferation and angiogenesis. Org Biomol Chem 2012; 10: 2979-2992.

24 

Folkman J, Langer R, Linhardt RJ, Haudenschild C, Taylor S. Angiogenesis inhibition and tumor regression caused by heparin or a heparin fragment in the presence of cortisone. Science 1983; 221: 719-725.

25 

Ma J, Waxman DJ. Combination of antiangiogenesis with chemotherapy for more effective cancer treatment. Mol Cancer Ther 2008; 7: 3670-3684.

26 

Griffith EC, Su Z, Turk BE, et al. Methionine aminopeptidase (type 2) is the common target for angiogenesis inhibitors AGM-1470 and ovalicin. Chem Biol 1997; 4: 461-471.

27 

Sin N, Meng L, Wang MQ, Wen JJ, Bornmann WG, Crews CM. The anti-angiogenic agent fumagillin covalently binds and inhibits the methionine aminopeptidase, MetAP-2. Proc Natl Acad Sci U S A 1997; 94: 6099-6103.

28 

Marino JP Jr, Fisher PW, Hofmann GA, et al. Highly potent inhibitors of methionine aminopeptidase-2 based on a 1,2,4-triazole pharmacophore. J Med Chem 2007; 50: 3777-3785.

29 

Sheppard GS, Wang J, Kawai M, et al. Discovery and optimization of anthranilic acid sulfonamides as inhibitors of methionine aminopeptidase-2: a structural basis for the reduction of albumin binding. J Med Chem 2006; 49: 3832-3849.

30 

Yeh JR, Mohan R, Crews CM. The antiangiogenic agent TNP-470 requires p53 and p21CIP/WAF for endothelial cell growth arrest. Proc Natl Acad Sci U S A 2000; 97: 12782-12787.

31 

Zhang Y, Griffith EC, Sage J, Jacks T, Liu JO. Cell cycle inhibition by the anti-angiogenic agent TNP-470 is mediated by p53 and p21WAF1/CIP1. Proc Natl Acad Sci U S A 2000; 97: 6427-6432.

32 

Brooks CL, Gu W. How does SIRT1 affect metabolism, senescence and cancer? Nat Rev Cancer 2009; 9: 123-128.

33 

Liu T, Liu PY, Marshall GM. The critical role of the class III histone deacetylase SIRT1 in cancer. Cancer Res 2009; 69: 1702-1705.

34 

Ota H, Akishita M, Eto M, Iijima K, Kaneki M, Ouchi Y. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol 2007; 43: 571-579.

35 

Shim JS, Matsui Y, Bhat S, et al. Effect of nitroxoline on angiogenesis and growth of human bladder cancer [published correction appears in J Natl Cancer Inst 2011; 103: 1070]. J Natl Cancer Inst 2010; 102: 1855-1873.

36 

Ramos A, Sadeghi S, Tabatabaeian H. Battling chemoresistance in cancer: root causes and strategies to uproot them. Int J Mol Sci 2021; 22: 9451.

37 

Gupta SK, Singh P, Ali V, Verma M. Role of membrane-embedded drug efflux ABC transporters in the cancer chemotherapy. Oncol Rev 2020; 14: 448.

38 

König J, Müller F, Fromm MF. Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacol Rev 2013; 65: 944-966.

39 

Levatić J, Ćurak J, Kralj M, Šmuc T, Osmak M, Supek F. Accurate models for P-gp drug recognition induced from a cancer cell line cytotoxicity screen. J Med Chem 2013; 56: 5691-5708.

40 

Li S, Zhang W, Yin X, et al. Mouse ATP-binding cassette (ABC) transporters conferring multi-drug resistance. Anticancer Agents Med Chem 2015; 15: 423-432.

41 

Choi YH, Yu AM. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr Pharm Des 2014; 20: 793-807.

42 

He C, Sun Z, Hoffman RM, et al. P-glycoprotein overexpression is associated with cisplatin resistance in human osteosarcoma. Anticancer Res 2019; 39: 1711-1718.

43 

Ji L, Liu X, Zhang S, et al. The novel triazolonaphthalimide derivative LSS-11 synergizes the anti-proliferative effect of paclitaxel via STAT3-dependent MDR1 and MRP1 downregulation in chemoresistant lung cancer cells. Molecules 2017; 22: 1822.

44 

Zhang X, Xiao W, Wang L, Tian Z, Zhang J. Deactivation of signal transducer and activator of transcription 3 reverses chemotherapeutics resistance of leukemia cells via down-regulating P-gp. PLoS One 2011; 6: e20965.

45 

Wagenlehner FM, Münch F, Pilatz A, et al. Urinary concentrations and antibacterial activities of nitroxoline at 250 milligrams versus trimethoprim at 200 milligrams against uropathogens in healthy volunteers. Antimicrob Agents Chemother 2014; 58: 713-721.

46 

Knowles M, Dyrskjøt L, Heath EI, Bellmunt J, Siefker-Radtke AO. Metastatic urothelial carcinoma. Cancer Cell 2021; 39: 583-585.

47 

Bhumsuk K, Section VI. Chemotherapy for metastatic bladder cancer, bladder cancer. Ja Hyeon Ku (eds.). Academic Pres 2018; 523-537.

48 

Xu N, Lin W, Sun J, et al. Nitroxoline inhibits bladder cancer progression by reversing EMT process and enhancing anti-tumor immunity. J Cancer 2020; 11: 6633-6641.

49 

Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2002; 2: 442-454.

50 

Zhao R, Trainor PA. Epithelial to mesenchymal transition during mammalian neural crest cell delamination. Semin Cell Dev Biol 2023; 138: 54-67.

51 

Cancer of the prostate - cancer stat facts. SEER. Available at: https://seer.cancer.gov/statfacts/html/prost.html (accessed: 28.03.2024).

52 

Prostate cancer - statistics. Cancer. Net. 2012. Available at: https://www.cancer.net/cancer-types/prostate-cancer/statistics (accessed: 28.03.2024).

53 

Kelly SP, Anderson WF, Rosenberg PS, Cook MB. Past, current, and future incidence rates and burden of metastatic prostate cancer in the United States. Eur Urol Focus 2018; 4: 121-127.

54 

James ND, Spears MR, Clarke NW, et al. Survival with newly diagnosed metastatic prostate cancer in the “Docetaxel Era”: data from 917 patients in the control arm of the STAMPEDE trial (MRC PR08, CRUK/06/019). Eur Urol 2015; 67: 1028-1038.

55 

Wu JN, Fish KM, Evans CP, Devere White RW, Dall’Era MA. No improvement noted in overall or cause-specific survival for men presenting with metastatic prostate cancer over a 20-year period. Cancer 2014; 120: 818-823.

56 

Ojuka EO. Role of calcium and AMP kinase in the regulation of mitochondrial biogenesis and GLUT4 levels in muscle. Proc Nutr Soc 2004; 63: 275-278.

57 

Russo GL, Russo M, Ungaro P. AMP-activated protein kinase: a target for old drugs against diabetes and cancer. Biochem Pharmacol 2013; 86: 339-350.

58 

Stolz A, Ertych N, Bastians H. Tumor suppressor CHK2: regulator of DNA damage response and mediator of chromosomal stability. Clin Cancer Res 2011; 17: 401-405.

59 

Antoni L, Sodha N, Collins I, Garrett MD. CHK2 kinase: cancer susceptibility and cancer therapy–two sides of the same coin? Nat Rev Cancer 2007; 7: 925-936.

60 

Zhang J, Willers H, Feng Z, et al. Chk2 phosphorylation of BRCA1 regulates DNA double-strand break repair. Mol Cell Biol 2004; 24: 708-718.

61 

Jack MT, Woo RA, Hirao A, Cheung A, Mak TW, Lee PW. Chk2 is dispensable for p53-mediated G1 arrest but is required for a latent p53-mediated apoptotic response. Proc Natl Acad Sci U S A 2002; 99: 9825-9829.

62 

Tanida I, Ueno T, Kominami E. LC3 and autophagy. Methods Mol Biol 2008; 445: 77-88.

63 

Repac Antić D, Parčina M, Gobin I, Petković Didović M. Chelation in antibacterial drugs: from nitroxoline to cefiderocol and beyond. Antibiotics (Basel) 2022; 11: 1105.

64 

Xu N, Huang L, Li X, et al. The novel combination of nitroxoline and PD-1 blockade, exerts a potent antitumor effect in a mouse model of prostate cancer. Int J Biol Sci 2019; 15: 919-928.

65 

Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res 2020; 10: 727-742.

66 

Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol 2007; 8: 239-245.

67 

Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci Transl Med 2016; 8: 328rv4.

68 

Thompson RH, Kuntz SM, Leibovich BC, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res 2006; 66: 3381-3385.

69 

Lyford-Pike S, Peng S, Young GD, et al. Evidence for a role of the PD-1: PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res 2013; 73: 1733-1741.

70 

Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S. Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother 2007; 56: 1173-1182.

71 

Gevensleben H, Dietrich D, Golletz C, et al. The immune checkpoint regulator PD-L1 is highly expressed in aggressive primary prostate cancer. Clin Cancer Res 2016; 22: 1969-1977.

72 

Rassy E, Boussios S, Pavlidis N. Genomic correlates of response and resistance to immune checkpoint inhibitors in carcinomas of unknown primary. Eur J Clin Invest 2021; 51: e13583.

73 

Biller LH, Schrag D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA 2021; 325: 669-685.

74 

Shi Y, Lei Y, Liu L, et al. Integration of comprehensive genomic profiling, tumor mutational burden, and PD-L1 expression to identify novel biomarkers of immunotherapy in non-small cell lung cancer. Cancer Med 2021; 10: 2216-2231.

75 

Chen W, Zhang H, Chen Z, et al. Development and evaluation of a novel series of Nitroxoline-derived BET inhibitors with antitumor activity in renal cell carcinoma. Oncogenesis 2018; 7: 83.

Copyright: © 2024 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.