215 REVIEW ARTICLE Possible use of antimicrobial agents in cancer therapy Copyright (c) 2022 Slovenian Medical Journal. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License. Possible use of antimicrobial agents in cancer therapy Možnosti uporabe protimikrobnih učinkovin pri zdravljenju raka Ana Mitrović,1 Janko Kos1,2 Abstract Cancer represents a major burden for health systems and is the second leading cause of death in the developed world. Despite major progress during past years, a lot of resources are still directed towards the identification of new antitumour drugs that would allow more efficient treatment, and prolonged patients’ survival, lower cancer recurrence rate, and de- creased side effects associated with cancer therapy, thus increased quality of life for cancer patients. However, the intro- duction of new drugs to clinical practice is associated with high costs. To keep the health system sustainable, the search for new indications of existing drugs has attracted a lot of attention in the field of oncology. Among others, antitumour activity has been shown for some antibiotics and other antimicrobial agents, including doxycycline, chloroquine, nitroxoline, and certain fluoroquinolones. In this review, we summarize various molecular mechanisms and tumour models used to define their antitumour activity. The latter has been demonstrated in a number of independent studies both in vitro as well as in vivo. Moreover, structures of existing compounds were used as lead compounds for the development of new derivatives and the identification of structural elements that improve antitumour activity. Together, these studies point to new possi- bilities for the use of already well-established drugs, which could expand their range, and, either, alone or in combination with existing therapy, improve the effectiveness of antitumour therapy. Izvleček Rak je veliko breme za zdravstvene sisteme in še vedno drugi najpogostejši vzrok smrti v razvitem svetu. Zato se, kljub velikem napredku v zadnjih letih, še vedno veliko sredstev usmerja v iskanje novih učinkovitejših protitumorskih zdravil, ki bi omogočila bolj učinkovito zdravljenje, daljše preživetje bolnikov z rakom, preprečila ponovni pojav raka in zmanjšala stranske učinke, povezane z zdravljenjem raka, s tem pa povečala kakovost življenja bolnikov z rakom. Uvajanje novih zdravil v klinično uporabo pa je hkrati povezano z vse večjimi stroški. Zato je zaradi velike potrebe po prepoznavanju novih protitumorskih zdravil iskanje novih indikacij za že obstoječa zdravila na področju onkologije pritegnilo veliko pozornosti. Pri tem se je med drugim protitumorsko delovanje pokazalo tudi za nekatere antibiotike in druge protimikrobne učinko- vine. Slednje se je med drugim izkazalo za doksiciklin, klorokin, nitroksolin in nekatere predstavnike fluorokinolonov. V 1 Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia 2 Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia Correspondence / Korespondenca: Ana Mitrović, e: ana.mitrovic@ijs.si Key words: drug repurposing; antitumour therapy; antimicrobial agents; cancer; drug discovery Ključne besede: premeščanje zdravil; protitumorska terapija; protimikrobne učinkovine; rak; iskanje novih učinkovin Received / Prispelo: 2. 11. 2020 | Accepted / Sprejeto: 16. 3. 2021 Cite as / Citirajte kot: Mitrović A, Kos J. Possible use of antimicrobial agents in cancer therapy. Zdrav Vestn. 2022;91(5–6):215–25. DOI: https://doi.org/10.6016/ZdravVestn.3188 eng slo element en article-lang 10.6016/ZdravVestn.3188 doi 2.11.2020 date-received 16.3.2021 date-accepted Cytology, oncology, cancerology Citologija, onkologija, kancerologija discipline Review article Pregledni znanstveni članek article-type Possible use of antimicrobial agents in cancer therapy Možnosti uporabe protimikrobnih učinkovin pri zdravljenju raka article-title Possible use of antimicrobial agents in cancer therapy Možnosti uporabe protimikrobnih učinkovin pri zdravljenju raka alt-title drug repurposing, antitumour therapy, antimi- crobial agents, cancer, drug discovery premeščanje zdravil, protitumorska terapija, protimikrobne učinkovine, rak, iskanje novih učinkovin kwd-group The authors declare that there are no conflicts of interest present. Avtorji so izjavili, da ne obstajajo nobeni konkurenčni interesi. conflict year volume first month last month first page last page 2022 91 5 6 215 225 name surname aff email Ana Mitrović 1 ana.mitrovic@ijs.si name surname aff Janko Kos 1,2 eng slo aff-id Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia Odsek za biotehnologijo, Inštitut Jožef Stefan, Ljubljana, Slovenija 1 Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia Fakulteta za farmacijo, Univerza v Ljubljani, Ljubljana, Slovenija 2 Slovenian Medical Journallovenian Medical Journal 216 CYTOLOGY, ONCOLOGY, CANCEROLOGY Zdrav Vestn | May – June 2022 | Volume 91 | https://doi.org/10.6016/ZdravVestn.3188 1 Introduction After cardiovascular diseases, cancer is one of the biggest health problems and the second leading cause of death in the developed world, including Slovenia. Ac- cording to the World Health Organization (WHO), 9.6 million people died of cancer worldwide in 2018 (1). In addition to being a major health problem, cancer also presents a major social and economic burden in today’s society (1). Despite major advances in cancer treatment in recent years, we still face many challenges (2). The key challenges in developing more effective therapies are to prevent cancer recurrence, avoid therapy resistance and reduce the many side-effects associated with most of the therapeutic approaches currently used in clinical prac- tice, which further reduce the quality of life for cancer patients (3). The development of new drugs is extreme- ly costly and the costs only continue to increase, which is why the number of new drugs on the market has de- clined over the last decade, despite high investment in research and development (4). In response to these high costs and the small num- ber of drugs that successfully pass all stages of testing and reach the market, more and more attention is being dedicated to finding new indications and uses for drugs already on the market (so-called drug repurposing, or repositioning) (5,6). The toxicological profiles, tolerabil- ity, pharmacokinetic and pharmacodynamic properties of these drugs are already well known, and these are the main advantages that increase their potential for use in cancer treatment. For existing drugs, their dosage regi- men, pharmacological properties and their interactions with other drugs are known (2,5-7). Due to the large amount of known data, the use of existing active sub- stances for new indications is therefore associated with shorter time and lower costs required for the substances’ successful translation into clinical use (2,5,6). When looking for new indications for existing drugs, the first step is to investigate in detail their mechanism of action on new targets and to test their efficacy and safety in clinical studies, as the use of a drug for a new indication may be associated with new side-effects that are not yet known (5,6). However, for existing drugs, it preglednem članku povzemamo različne molekulske mehanizme, uporabljene tumorske modele in raziskave, ki oprede- ljujejo njihovo protitumorsko delovanje. To so prikazale številne neodvisne študije tako in vitro kot in vivo, strukture ob- stoječih učinkovin pa so se hkrati uporabile kot spojine za razvoj novih derivatov in identificiranje strukturnih elementov, ki izboljšajo protitumorsko delovanje. Te raziskave tako kažejo na nove možnosti za uporabo že dobro znanih učinkovin, ki bi lahko razširile svoj spekter uporabe in samostojno ali v kombinaciji z že obstoječim zdravljenjem izboljšale uspešnost protitumorskega zdravljenja. is possible to move more quickly into phases 2 and 3 of clinical trials due to the large amount of information al- ready obtained in previous pre-clinical and clinical trials (2). In addition to drugs that are in clinical use, active substances that have been shown to be safe in clinical trials but have not entered clinical use for other reasons are also well suited to the search for new indications (6). The drug repurposing has attracted particular atten- tion in the field of oncology, due to the great need to identify new, more effective active substances (3,5). It is now known that many active substances that are used to treat other indications and differ in both their structure and mechanism of action, can also show antitumour ac- tivity. Antitumour activity has already been confirmed for thalidomide, metformin, acetylsalicylic acid, statins, raloxifene, tamoxifen, certain antidepressants and anti- psychotics, and many other drugs (2,5). In addition to these, some antibiotics and antimi- crobials have also shown antitumour activity, including doxycycline, chloroquine, nitroxoline and fluroquinolo- nes (Figure 1). In this review article, we will focus on the potential use of some antimicrobial agents for cancer treatment and present possible mechanisms for their an- titumour activity. 2 Doxycycline Doxycycline (Figure 1) is a tetracycline antibiotic used to treat a wide range of bacterial infections (5,8). It is also used prophylactically in malaria prevention and is effective in the treatment of malaria in combination with quinine. Doxycycline is rapidly absorbed and rapidly crosses the blood-brain barrier following oral adminis- tration, its pharmacokinetic properties and half-life are known, and it is generally well tolerated by patients fol- lowing administration (8). In recent years, several studies have shown that, in addition to its potent antibacterial activity, doxycycline has antitumour activity. Antitumour activity of doxycy- cline has been demonstrated in various types of cancer, including breast, cervical, ovarian, prostate, lung, oral, 217 REVIEW ARTICLE Possible use of antimicrobial agents in cancer therapy Figure 1: Structures of antimicrobial agents exhibiting antitumour activity. N OH NO 2 OH O OH O O NH 2 OH H OH N OH H NCl HN N X NR 7 F R 5 O OH O R 2 R 1 NN F O OH O HN NN F O OH O HN F NH 2 N NN F O OH O H2N N O N NN F O OH O HN Doxycycline Fluoroquinolones Chloroquine Nitroxoline General structure of fluoroquinolones Ciprofloxacin Enoxacin Gemifloxacin Sparfloxacin Selected fluoroquinolones with antitumor activity pancreatic and duodenal cancer, colorectal cancer, mel- anoma and leukaemia (5,8). More than 30 years ago, in leukaemia cells, doxycycline treatment was shown to inhibit T-cell proliferation and lead to their complete destruction, indicating that the mechanism of action of doxycycline depends on its concentration and the de- gree of tumour progression (9). Several mechanisms by which doxycycline inhibits tumour formation and progression have been described (8). As one of its mechanisms of action, doxycycline in- hibits the matrix metallopeptidases MMP-2 and MMP- 9, which are important players in various processes of cancer onset and progression. It further inhibits MMP activity by increasing the expression of tissue inhibitor MMP-2 (TIMP-2) (5,10-12). In animal models, doxy- cycline has been shown to inhibit metastases by inhib- iting MMP-2 and MMP-9 (5,13,14). Furthermore, dox- ycycline inhibits cancer progression through a number of signalling pathways (5,8). Among others, it directly inhibits protease-activated receptor 1 (PAR-1) signalling and downregulates VEGF signalling (10,15,16). Doxycy- cline also inhibits tumour cell adhesion, migration and invasion by inhibiting the expression and phosphoryla- tion of adhesion molecules such as focal adhesion kinase (FAK) in leukaemia cells and melanoma cells (11,12). It also reduces angiogenesis and cancer cell migration by inhibiting the expression of interleukin 8 (IL-8) (17). By inhibiting doxycycline signalling pathways, it inhibits tumour cell invasion, epithelial-mesenchymal transition of tumour cells and metastasis formation (5,8,14). Doxycycline induces apoptosis and inhibits cell pro- liferation (8,17). Inhibition of apoptosis involves both the mitochondria-dependent and caspase-dependent pathways (8,18). Doxycycline has been shown to induce cell cycle arrest in the G2/M phase in prostate cancer cells (19). According to the results in different types of cancer, doxycycline promotes apoptosis and inhibits cell proliferation by inhibiting various molecular targets that 218 CYTOLOGY, ONCOLOGY, CANCEROLOGY Zdrav Vestn | May – June 2022 | Volume 91 | https://doi.org/10.6016/ZdravVestn.3188 are importantly involved in these processes (8). Doxycycline is as an antitumour agent of particular interest for its action against cancer stem cells (8). In cer- vical cancer, doxycycline has been shown to reduce the expression of cancer stem cell markers such as SOX-2, OCT-4, NANOG and NOTCH (20). The anti-tumour activity of doxycycline has also been evaluated in clinical studies. However, in a phase 2 clini- cal study in patients with breast cancer and bone metas- tases, doxycycline did not show significant effects (21), while in a small pilot clinical study involving 15 patients, it showed a positive effect in almost 90% of persons with early forms of breast cancer, who received doxycycline 14 days prior to surgery. The expression of cancer stem cell markers (22) decreased in tumour samples following doxycycline treatment, showing promising possibilities for its use in cancer therapy. However, this needs to be further evaluated in a larger clinical study. Doxycycline is also effective in combination with oth- er known chemotherapeutic agents, making it a promis- ing active substance already in clinical use that could be used in oncology to treat cancer alone or in combination with conventional therapy to treat cancer and prevent its recurrence (8). 3 Chloroquine Chloroquine (7-Chloro-4-(4-Diethylamino-1-Meth- ylbutylamino)-Quinoline) (Figure 1) and its analogue hydroxychloroquine are used for the prevention and treatment of malaria (23-25). Chloroquine is an old drug, synthesised as early as 1934. It was used as the compound of choice in the treatment of malaria until the emergence of resistant strains (24). Later, its use was extended to the treatment of rheumatoid arthritis and lupus erythematosus (23,25). Chloroquine’s anti- tumour activity was first observed in the 1970s when, during a study of its effect against malaria, the incidence of Burkitt’s lymphoma was reduced in a group receiving chloroquine. However, it was only later that the latter attracted more interest (26). To date, the antitumour ac- tivity of chloroquine and hydroxychloroquine has been demonstrated in several in vitro and in vivo studies in mouse models with different types of cancer, including breast, liver and colon cancer, glioblastoma and melano- ma. It reduced tumour progression and slowed tumour growth (25). In a mouse model of liver cancer, a reduc- tion in the number and size of metastases in the lungs was also shown (27). As antitumour agents, chloroquine and hydroxy- chloroquine can be used individually or as supportive therapies to improve the effect of chemotherapy and ra- diotherapy in different types of cancer. They are current- ly involved in several clinical studies (23-25). As of Feb- ruary 2021, 21 clinical trials for chloroquine in cancer and 89 clinical trials for hydroxychloroquine have been registered on clinicaltrials.gov, which are at various stag- es, many of the trials still ongoing, with results available for only some of the trials (28). In clinical trials, patients mainly received chloroquine and hydroxychloroquine in combination with other antitumour agents (25,28). Results to date show that for some types of cancer, pa- tients receiving chloroquine or hydroxychloroquine as supportive therapy have prolonged survival compared to the control group (25,29). A significant improvement in survival of patients receiving chloroquine in addition to radiotherapy and chemotherapy was already observed in one of the first clinical trials, which started in May 1998, in 18 patients with glioblastoma (30). A similar improvement in survival rate of glioblastoma patients who received chloroquine in addition to conventional chemotherapy and radiation, when compared to those who received placebo, was also observed in a clinical study conducted in October 2000. The study included 15 patients in each group (31). The retrospective study pooled all data obtained over a 5-year period from 41 glioblastoma patients who received chloroquine as ad- juvant therapy and were not included in the aforemen- tioned study (31). In two independent phase 2 clinical studies (32) and a prospective study, in one group (33) of patients with brain metastases, chloroquine was shown to increase the sensitivity of cells to radiotherapy in pa- tients with brain metastases. It prolongs both the metas- tasis progression-free survival and overall survival, sug- gesting the possibility of its use as a supportive treatment for radiation therapy (25). Furthermore, several clinical studies of different types of cancer have shown that the addition of hydroxychloroquine to conventional therapy improves response to therapy by inhibiting autophagy (25). Due to their long-standing use in clinical practice, the side-effects of chloroquine and hydroxychloroquine are well known. Serious side effects rarely occur after their short-term use. However, adverse toxic effects, in particular nephrotoxicity, may occur after prolonged use at higher doses and when taken concomitantly with other antitumour agents (23,25,34). The presence of an additional hydroxy group in hydroxychloroquine sig- nificantly reduces toxicity and affects the pharmacoki- netic properties of the compound, while the differences in antitumour activity of chloroquine and hydroxychlo- roquine are not fully known (25,35). 219 REVIEW ARTICLE Possible use of antimicrobial agents in cancer therapy Chloroquine and hydroxychloroquine are highly sol- uble and are rapidly and well absorbed after oral admin- istration (24,25). Chloroquine is a weak base; at physio- logical pH 7.4, it is found in an unprotonated or partially protonated form, which allows it to pass well through cell membranes and enter acidic cell compartments. As a weak base, chloroquine is protonated and retained in lysosomes after entering the acidic environment of ly- sosomes. This also results in an increase in the pH of the lysosomes, which reduces their function, as most of the proteins in lysosomes have their optimum activity at acidic pH (23-25). The retention of chloroquine inside cells leads to inhibition of autophagy, one of the key mechanisms of its antitumour activity (23-25,34). Chloroquine and hydroxychloroquine inhibit autophagy by inhibiting autophagosome fusion and degradation within lyso- somes due to an increase in pH (25,36). It also affects autophagy via the PI3K/Akt/mTOR signalling path- way, where it has a synergistic effect with inhibitors of the AMPK signalling pathway and the activated Janus kinase-2 (JAK2)/STAT3 signalling pathway (23,25,37). In addition to early autophagy, chloroquine also inhibits late stages of autophagy and can induce cell death even under conditions where inhibitors of early stages of au- tophagy cannot (23). Several independent studies have also shown an antitumour activity of chloroquine, independent of in- hibition of autophagy (24). One of the mechanisms is modulation of cellular metabolism, affecting amino ac- id metabolism, glucose metabolism and mitochondrial metabolism (23,24). Chloroquine and hydroxychloro- quine also affect apoptosis and cell cycle arrest by regu- lating molecules importantly involved in these processes (25). Moreover, chloroquine can also induce cell death via lysosome-mediated apoptosis by acting on cathep- sins (24). Chloroquine is also involved in the regula- tion of cellular stress and modulators of inflammation by regulating proteostasis via the ubiquitin/proteasome system (23). Next, it affects cell proliferation and surviv- al by acting on glutamate dehydrogenase activity (25). Chloroquine also exerts antitumour effects by normalis- ing tumour vasculature by reducing vascular density and increasing tight junction formation (25,38). Several molecular targets of the antitumour activity of chloroquine and hydroxychloroquine are known, in- cluding the NF-κB transcription factor, p53 tumour sup- pressor factor and CXCL12/CXCR4 signalling pathways (24,25). By inhibiting the CXCL12/CXCR4 signalling pathway, chloroquine reduces phosphorylation of the signalling molecules ERK and STAT3 (25) and thereby also acts on cancer stem cells (23). Additionally, in a bile duct cancer cell line, chloroquine inhibits tumour cell metastasis by affecting hypoxia-inducible factor 1α (HIF-1α), VEGF and the epithelial-mesenchymal tran- sition process (25). By inhibiting autophagy, chloroquine can bypass the limitations of existing chemotherapy, as autophagy is, among other things, one of the key processes for sur- vival and resistance to existing chemotherapy in cancer stem cells and facilitates the epithelial-mesenchymal transition of tumour cells. This is one of the process- es by which cells can acquire the properties of cancer stem cells (23,25). In cancer stem cells, chloroquine at a concentration of 20 μM was shown to affect stemness by specific action on autophagy (23,39). It was further shown that in cancer stem cells, low micromolar con- centrations of chloroquine affect JAK2/STAT3, Hedge- hog and CXC4 signalling pathways, DNA methyl trans- ferase expression and expression of stemness markers (23,25,40). Chloroquine has also been shown in several studies to be involved in regulating the immune system and the inflammatory response through various mech- anisms (23,24). When in concomitant use, chloroquine improves the effect of chemotherapy and radiotherapy. By inhibiting autophagy, it increases the sensitivity of tumour cells to radiotherapy, which has been shown in various types of cancer including breast cancer, lung cancer, and glioma. A meta-analysis of studies has shown that addition of chloroquine to chemotherapy or radiotherapy increases patient survival and prolongs disease-free survival (23). Chloroquine and hydroxychloroquine have a synergis- tic effect with BET inhibitors already at low concentra- tions and induce cancer stem cell apoptosis in pancreatic cancer and acute myeloid leukaemia (23,41,42). Chlo- roquine also increases the efficiency and killing of tu- mour cells in combination with cell cycle inhibitors and leads to apoptosis together with proteasome inhibitors, as shown in a mouse model of liver cancer (23,43). Fur- thermore, inhibitors of the AMPK signalling pathway and the STAT3 signalling pathway have also been shown to have a synergistic effect on cell death (23). Despite promising results from pre-clinical studies, chloroquine significant antitumour effect in clinical studies was not yet shown. Key reasons for this could be that chloroquine is less able to enter tumour cells due to the acidic extracellular environment of tumours, and that chloroquine has an important role in regulating the immune response by inhibiting autophagy in lyso- somes (23,25). In addition, the different dependence of tumours on autophagy results in different responses to 220 CYTOLOGY, ONCOLOGY, CANCEROLOGY Zdrav Vestn | May – June 2022 | Volume 91 | https://doi.org/10.6016/ZdravVestn.3188 chloroquine therapy in different types of tumours (25). At the same time, it was shown that high concentrations of chloroquine are required to achieve a therapeutic ef- fect on autophagy inhibition in leukaemia, which lim- its its use. Nevertheless, chloroquine has been shown to affect metastatic tumour cells that are not in an acidic tumour environment and are particularly sensitive to lysosomal inhibition. This reduces the possibility of me- tastasis formation (23). In triple-negative breast cancer, chloroquine successfully kills cancer stem cells and re- duces the ability of tumours to metastasise in both in vitro and in vivo mouse models (44). Chloroquine has been shown to act primarily on tumour cells with can- cer stem cell properties. Therefore, its use is particularly appropriate in the context of concomitant use of agents targeting differentiated tumour cells (23,25). 4 Nitroxoline Nitroxoline (5-nitro-8-hydroxyquinoline) (Figure 1) is a well-known antimicrobial agent used in the treat- ment of urinary tract infections. Nitroxoline’s antimicro- bial activity was discovered in the 1950s and it was fast used in clinical practice (45). Despite it being used for a long time, bacteria have still not developed resistance to nitroxoline (45,46). Nitroxoline is effective against most Gram-positive and Gram-negative pathogenic uri- nary tract bacteria, mycoplasmas (M. hominis, Ureaplas- ma urealyticum) and human pathogens Candida spp. (45). In addition, nitroxoline showed efficacy against most other Gram-negative bacteria and was also effec- tive against Gram-positive bacteria, suggesting poten- tial use as an antimicrobial agent for other indications in addition to the treatment of urinary tract infections (47). Nitroxoline exerts its antimicrobial activity mainly by chelation of various bivalent ions (45,48,49). In this way, it stabilises lipopolysaccharides on the bacterial surface, which increases the hydrophobicity of the bac- terial surface and reduces their adhesion (45,48,49). By this nitroxoline also inhibits RNA synthesis during yeast cell division (45,50), while at lower concentrations, it in- hibits the formation of bacterial biofilms (45,46,51). As an established antimicrobial agent, nitroxoline has well- known pharmacokinetic and pharmacodynamic prop- erties (45,52-54). Nitroxoline is completely and rapidly absorbed in the urinary tract after oral administration (52,54), and only minor and tolerable side effects have been observed, suggesting that nitroxoline administra- tion is safe (45). Antitumour activity of nitroxoline has been demon- strated by several studies to date. Its antitumour activity was first identified in the search for methionine amino- peptidase type 2 (MetAP2) inhibitors, which could be used as new anti-angiogenic agents (55). Nitroxoline has been shown as potent inhibitor of MetAP2 in vitro, in- hibiting endothelial cell proliferation and angiogenesis both in vitro and in vivo. In addition, nitroxoline induces senescence and inhibits HUVEC endothelial cell prolif- eration by simultaneously inhibiting MetAP2 and Sir- tuin 1 (Sirt1). By Sirt1 inhibition, nitroxoline increases the amount of acetylated tumour suppressor p53, result- ing in reduced angiogenesis. At the same time, nitrox- oline significantly reduced the size of breast tumours in a mouse model and inhibited the growth of bladder cancer in an orthotopic mouse model (55). Nitroxoline was later shown to reduce the expression of proteins as- sociated with the epithelial-mesenchymal transition of tumour cells and the amount of myeloid-derived sup- pressor cells (MDSCs) in peripheral blood in bladder cancer (56). Furthermore, nitroxoline was identified in a second, independent study by high throughput com- pound library search and biochemical evaluation of the best hits as a potent, reversible and non-covalent in- hibitor of the lysosomal cysteine peptidase cathepsin B (57), which plays an important role in the initiation and progression of cancer, its invasion and metastasis, and is known to be a promising target for the development of novel antitumour agents (58-60). The crystal structure of the nitroxoline-cathepsin B complex shows that nitroxo- line binds to the active site cleft of cathepsin B and there- by selectively inhibits its endopeptidase activity at low micromolar concentrations (57). In various functional assays in cell lines and murine tumour models, nitrox- oline significantly reduced extracellular matrix degra- dation, tumour cell invasion, metastasis and endothelial tube formation in an in vitro model of angiogenesis (58). Nitroxoline acts as an antitumour agent and inhibits tumour cell migration by reducing the expression of the tumour transcription factor FOXM1, an important reg- ulator of cancer progression. In the same study, nitroxo- line was also shown to reduce the expression of MMP-2 and MMP-9, which are important players in tumour mi- gration and invasion and whose expression is controlled by, among others, FOXM1. In addition to inhibition of MMP-2 and MMP-9 by inhibiting FOXM1, molecular interaction studies suggest that nitroxoline also direct- ly interacts with the catalytic domains of MMP-2 and MMP-9 (61). Furthermore, several studies have shown that nitrox- oline inhibits tumour cell proliferation. This has been shown in cells from a variety of cancer types, includ- ing malignant glioma, multiple myeloma, glioblastoma, 221 REVIEW ARTICLE Possible use of antimicrobial agents in cancer therapy leukaemia, prostate cancer, bile duct cancer (cholangio- carcinoma), pancreatic cancer, small cell lung cancer and bladder cancer (56,62). The antitumour activity of nitroxoline has been shown to be dose-dependent, as well as dependent on the period of administration, with lower doses leading mainly to cell cycle arrest in the G0/ G1 phase, while higher doses promote apoptosis. Ni- troxoline promotes apoptosis and cell cycle arrest in the G0/G1 phase by acting on various molecular targets that are importantly involved in these processes (62). The an- titumour activity of nitroxoline on pancreatic cell lines was further improved when nitroxoline was co-adminis- tered together with nelfinavir, a competitive inhibitor of HIV aspartate peptidase, and other antiviral agents for the treatment of HIV infection (63). However, according to data available at clinicaltrials.gov, the antitumour ac- tivity of nitroxoline has not yet been evaluated in clinical studies (28). To further improve the antitumour activity of nitrox- oline, several new derivatives with different structural modifications have been prepared by structure-based chemical synthesis, based on the structure of nitroxoline (62). By modifying various substituents, a large number of nitroxoline derivatives was prepared with the aim to improve cathepsin B inhibition (57,64-66), its anti-an- giogenic action by inhibiting MetAP2 and SIRT1 (67) and inhibition of BET proteins, which bind competitive- ly to BRD-BD1 and are involved in maintaining chro- matin stability, thereby controlling cell cycle progression (62,68,69). The preparation of new derivatives also re- vealed the structural features needed to improve activity on a particular target (62). Additionally, the antitumour activity of nitroxoline and the inhibition of cathepsin B were also improved by the preparation of organoru- thenium complexes with nitroxoline and its derivatives (70). The antitumour activity of nitroxoline, and in par- ticular its uptake into lysosomes, was also improved by its incorporation into nanoparticles composed of bovine serum albumin, copper ions and nitroxoline (BSA/Cu/ NQ) (71). Nitroxoline is a promising, known active substance that could be used to treat various types of cancer. Ni- troxoline is also an interesting compound for the devel- opment of new derivatives with improved antitumour activity and selectivity for specific targets. 5 Fluoroquinolones Fluoroquinolones (Figure 1) are the largest group of antimicrobial agents. Fluoroquinolones inhibit bacterial DNA duplication and transcription, either by inhibiting bacterial DNA gyrase or topoisomerase II (4). Nalidixic acid was discovered as the first quinolone with antimi- crobial activity in the 1960s. The discovery was followed by further development and optimization of quinolones. They can be divided into four generations based on their pharmacokinetic and antimicrobial properties. Initially, quinolones were only effective against Gram-negative bacteria, but as their structure has been optimized, their spectrum of activity has broadened. Fourth-generation fluoroquinolones are effective against Gram-negative and Gram-positive bacteria, anaerobes Pseudomonas sp. and atypical bacteria (72). In addition to their antibac- terial activity, some fluoroquinolones have been shown to be effective in combination with chloroquine in the treatment of malaria. They have also been shown to work against some other parasites, such as Trypanosoma brucei and Toxoplasma gondii (4). In addition to their direct effect on bacterial growth, fluoroquinolones con- tribute to antimicrobial activity by their immunomodu- latory properties. Fluoroquinolones, especially those of the newer generation such as ciprofloxacin, norfloxacin and ofloxacin, inhibit cytokine synthesis even at low concentrations. They have also displayed an anti-inflam- matory response and a protective role against lipopoly- saccharide-induced liver injury (4,73). Several independent studies have demonstrated an- titumour activity of fluoroquinolones, with multiple different mechanisms of antitumour action. Different fluoroquinolones have been shown to stop tumour pro- gression with cell cycle arrest, mainly in the S/G2 phase, and by inducing tumour cell apoptosis (4). Ciprofloxacin has been shown as the most potent inducer of apoptosis and has induced apoptosis in cells from a variety of can- cers, including prostate, bladder, pancreatic, colorectal cancer and others (4,74,75). In contrast, a more recent study of ciprofloxacin showed the opposite effect on can- cer progression, showing it promotes the formation of cells with a cancer stem cell phenotype in human small cell lung cancer cells. Here, ciprofloxacin was shown to increase the expression of stemness markers and pro- teins required for cell self-renewal (76). Among fluoro- quinolones, only gemifloxacin has been shown to inhibit tumour cell metastasis by inhibiting migration and inva- sion through inhibition of TNFα-stimulated NFκB acti- vation (77,78). Additionally, sparfloxacin was shown to have an impact on invasion and migration of colon can- cer tumour cells (79). Enoxacin, due to its unique struc- ture, shows antitumour effects by inhibiting the produc- tion of microRNA (miRNA), because it is oncogenic and contributes to tumour progression (4,80,81). Several in- dependent studies showed fluoroquinolones, including 222 CYTOLOGY, ONCOLOGY, CANCEROLOGY Zdrav Vestn | May – June 2022 | Volume 91 | https://doi.org/10.6016/ZdravVestn.3188 References 1. World Health Organization. Cancer. Geneva: WHO; 2020 [cited 2021 Jan 17]. Available from: https://www.who.int/news-room/fact-sheets/detail/ cancer. 2. Gupta SC, Sung B, Prasad S, Webb LJ, Aggarwal BB. Cancer drug discovery by repurposing: teaching new tricks to old dogs. Trends Pharmacol Sci. 2013;34(9):508-17. DOI: 10.1016/j.tips.2013.06.005 PMID: 23928289 3. Shim JS, Liu JO. Recent advances in drug repositioning for the discovery of new anticancer drugs. Int J Biol Sci. 2014;10(7):654-63. DOI: 10.7150/ ijbs.9224 PMID: 25013375 4. Yadav V, Talwar P. Repositioning of fluoroquinolones from antibiotic to anti-cancer agents: an underestimated truth. Biomed Pharmacother. 2019;111(111):934-46. DOI: 10.1016/j.biopha.2018.12.119 PMID: 30841473 ciprofloxacin, fleroxacin, moxifloxacin and enoxacin, improve the antitumour effects of chemotherapeutic agents when co-administered with existing chemother- apeutic agents used in clinical practice (4). Several clini- cal trials are also currently underway to test the anti-tu- mour activity of fluoroquinolones in combination with established therapies in different types of cancer, such as bladder cancer and acute myeloid leukaemia. The final results have yet to be published (28). The antitumour activity of fluoroquinolones can be further enhanced by the formation of complexes with metal ions. The latter was first demonstrated for com- plexes of norfloxacin with copper, which showed im- proved antitumour activity against leukaemia cells (82). This led to the preparation of metal complexes with oth- er fluoroquinolones, further improving the anti-tumour activity against different cancer cell lines compared to the initial compound. The gold-ruthenium complexes proved to be particularly effective in this regard, with an- titumour activity mainly against metastatic tumour cells, but without toxic effects on normal, healthy cells. The antitumour activity of ruthenium complexes is thought to be due to action on multiple targets, while the antitu- mour activity of gold complexes is thought to be mainly due to alteration of mitochondrial function and inhibi- tion of protein synthesis. Additionally, the introduction of nitrogen adducts into metal complexes with fluoro- quinolones inhibits their elimination from cells, thus al- lowing for increased retention of active compounds in tumours (4). Moreover, research on fluoroquinolones is also fo- cused on finding structural modifications to the mole- cules of existing fluoroquinolones that could further im- prove their antitumour activity. The presence of major functional groups at the C-7 site has been shown to be beneficial for enhancing antitumour activity (4,83). The large group of fluoroquinolones represents a promising group of antimicrobial drugs whose use could be extended to the treatment of cancer due to their an- titumour activity. At the same time, structural modifi- cations of existing fluoroquinolones could lead to the development of new molecules with improved antitu- mour activity. 6 Conclusion Successful cancer treatment still faces many chal- lenges. One of the important steps is to develop new drugs to avoid the limitations of current therapeutic approaches, such as resistance to therapy and the ma- ny side effects, associated with existing chemotherapeu- tics. However, the high costs associated with bringing a new drug to market have recently led to an increasing focus on finding new indications for existing drugs. It was shown that some antimicrobial drugs that have long been used successfully in clinical practice have antitu- mour activity. Among the latter, antitumour activity was also demonstrated for doxycycline, chloroquine, nitroxoline and fluoroquinolones that are discussed in detail in this review article. Their antitumour activity has been demonstrated in a number of tumour models and on different cancer types, and different mechanisms of antitumour action have been described. Known anti- microbial agents with antitumour activity also represent interesting compounds for the development of new de- rivatives with improved antitumour activity. Despite the numerous studies confirming the antitumour activity of anti-microbial agents, further clinical studies are needed to confirm their efficacy and allow their successful in- troduction into clinical practice. Attention should also be paid to possible interactions with existing therapies, in particular immune therapies and antibiotic therapy, which cancer patients often receive because of infections that result from their weakened immune system. Drug repurposing is therefore a promising approach that will allow faster and more efficient development of new an- ti-tumour therapies and improve the success of cancer treatment. Conflict of interest None declared. 223 REVIEW ARTICLE Possible use of antimicrobial agents in cancer therapy 5. Sleire L, Førde HE, Netland IA, Leiss L, Skeie BS, Enger PØ. Drug repurposing in cancer. Pharmacol Res. 2017;124:74-91. DOI: 10.1016/j. phrs.2017.07.013 PMID: 28712971 6. Hernandez JJ, Pryszlak M, Smith L, Yanchus C, Kurji N, Shahani VM, et al. Giving drugs a second chance: overcoming regulatory and financial hurdles in repurposing approved drugs as cancer therapeutics. Front Oncol. 2017;7(NOV):273. DOI: 10.3389/fonc.2017.00273 PMID: 29184849 7. Nowak-Sliwinska P, Scapozza L, Ruiz i Altaba A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim Biophys Acta Rev Cancer. 2019;1871(2):434-54. DOI: 10.1016/j.bbcan.2019.04.005 PMID: 31034926 8. Antoszczak M, Markowska A, Markowska J, Huczyński A. Old wine in new bottles: drug repurposing in oncology. Eur J Pharmacol. 2019;2020:866. PMID: 31730760 9. van den Bogert C, Dontje BH, Kroon AM. The antitumour effect of doxycycline on a T-cell leukaemia in the rat. Leuk Res. 1985;9(5):617-23. DOI: 10.1016/0145-2126(85)90142-0 PMID: 3874329 10. Wang SQ, Zhao BX, Liu Y, Wang YT, Liang QY, Cai Y, et al. New application of an old drug: antitumor activity and mechanisms of doxycycline in small cell lung cancer. Int J Oncol. 2016;48(4):1353-60. DOI: 10.3892/ ijo.2016.3375 PMID: 26846275 11. Wang C, Xiang R, Zhang X, Chen Y. Doxycycline inhibits leukemic cell migration via inhibition of matrix metalloproteinases and phosphorylation of focal adhesion kinase. Mol Med Rep. 2015;12(3):3374- 80. DOI: 10.3892/mmr.2015.3833 PMID: 26004127 12. Sun T, Zhao N, Ni CS, Zhao XL, Zhang WZ, Su X, et al. Doxycycline inhibits the adhesion and migration of melanoma cells by inhibiting the expression and phosphorylation of focal adhesion kinase (FAK). Cancer Lett. 2009;285(2):141-50. DOI: 10.1016/j.canlet.2009.05.004 PMID: 19482420 13. Shen LC, Chen YK, Lin LM, Shaw SY. Anti-invasion and anti-tumor growth effect of doxycycline treatment for human oral squamous-cell carcinoma—in vitro and in vivo studies. Oral Oncol. 2010;46(3):178-84. DOI: 10.1016/j.oraloncology.2009.11.013 PMID: 20036604 14. Qin Y, Zhang Q, Lee S, Zhong WL, Liu YR, Liu HJ, et al. Doxycycline reverses epithelial-to-mesenchymal transition and suppresses the proliferation and metastasis of lung cancer cells. Oncotarget. 2015;6(38):40667-79. DOI: 10.18632/oncotarget.5842 PMID: 26512779 15. Zhong W, Chen S, Zhang Q, Xiao T, Qin Y, Gu J, et al. Doxycycline directly targets PAR1 to suppress tumor progression. Oncotarget. 2017;8(10):16829-42. DOI: 10.18632/oncotarget.15166 PMID: 28187433 16. Zhong W, Chen S, Qin Y, Zhang H, Wang H, Meng J, et al. Doxycycline inhibits breast cancer EMT and metastasis through PAR-1/NF-κB/miR-17/ E-cadherin pathway. Oncotarget. 2017;8(62):104855-66. DOI: 10.18632/ oncotarget.20418 PMID: 29285218 17. Son K, Fujioka S, Iida T, Furukawa K, Fujita T, Yamada H, et al. Doxycycline induces apoptosis in PANC-1 pancreatic cancer cells. Anticancer Res. 2009;29(10):3995-4003. PMID: 19846942 18. Song H, Fares M, Maguire KR, Sidén Å, Potácová Z. Cytotoxic effects of tetracycline analogues (doxycycline, minocycline and COL-3) in acute myeloid leukemia HL-60 cells. PLoS One. 2014;9(12):e114457. DOI: 10.1371/journal.pone.0114457 PMID: 25502932 19. Zhu C, Yan X, Yu A, Wang Y. Doxycycline synergizes with doxorubicin to inhibit the proliferation of castration-resistant prostate cancer cells. Acta Biochim Biophys Sin (Shanghai). 2017;49(11):999-1007. DOI: 10.1093/ abbs/gmx097 PMID: 28985240 20. Yang B, Lu Y, Zhang A, Zhou A, Zhang L, Zhang L, et al. Doxycycline Induces Apoptosis and Inhibits Proliferation and Invasion of Human Cervical Carcinoma Stem Cells. PLoS One. 2015. ;10(6)DOI: 10.1371/ journal.pone.0129138 PMID: 226111245 21. Addison CL, Simos D, Wang Z, Pond G, Smith S, Robertson S, et al. A phase 2 trial exploring the clinical and correlative effects of combining doxycycline with bone-targeted therapy in patients with metastatic breast cancer. J Bone Oncol. 2016;5(4):173-9. DOI: 10.1016/j.jbo.2016.06.003 PMID: 28008379 22. Scatena C, Roncella M, Di Paolo A, Aretini P, Menicagli M, Fanelli G, et al. Doxycycline, an Inhibitor of Mitochondrial Biogenesis, Effectively Reduces Cancer Stem Cells (CSCs) in Early Breast Cancer Patients: A Clinical Pilot Study. Front Oncol. 2018;8:452. DOI: 10.3389/fonc.2018.00452 PMID: 30364293 23. Varisli L, Cen O, Vlahopoulos S. Dissecting pharmacological effects of chloroquine in cancer treatment: interference with inflammatory signaling pathways. Immunology. 2020;159(3):257-78. DOI: 10.1111/ imm.13160 PMID: 31782148 24. Weyerhäuser P, Kantelhardt SR, Kim EL. Re-purposing chloroquine for glioblastoma: potential merits and confounding variables. Front Oncol. 2018;8(AUG):335. DOI: 10.3389/fonc.2018.00335 PMID: 30211116 25. Verbaanderd C, Maes H, Schaaf MB, Sukhatme VP, Pantziarka P, Sukhatme V, et al. Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience. 2017;11:781. DOI: 10.3332/ecancer.2017.781 PMID: 29225688 26. Geser A, Brubaker G, Draper CC. Effect of a malaria suppression program on the incidence of African Burkitt’s lymphoma. Am J Epidemiol. 1989;129(4):740-52. DOI: 10.1093/oxfordjournals.aje.a115189 PMID: 2923122 27. Jiang PD, Zhao YL, Deng XQ, Mao YQ, Shi W, Tang QQ, et al. Antitumor and antimetastatic activities of chloroquine diphosphate in a murine model of breast cancer. Biomed Pharmacother. 2010;64(9):609-14. DOI: 10.1016/j.biopha.2010.06.004 PMID: 20888174 28. National Library of Medicine. ClinicalTrials.gov. Bethesda: US NLM; 2021 [cited 2021 Feb 13]. Available from: https://clinicaltrials.gov. 29. Xu R, Ji Z, Xu C, Zhu J. The clinical value of using chloroquine or hydroxychloroquine as autophagy inhibitors in the treatment of cancers: A systematic review and meta-analysis. Medicine (Baltimore). 2018;97(46):e12912. DOI: 10.1097/MD.0000000000012912 PMID: 30431566 30. Briceño E, Reyes S, Sotelo J. Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine. Neurosurg Focus. 2003;14(2):e3. DOI: 10.3171/foc.2003.14.2.4 PMID: 15727424 31. Sotelo J, Briceño E, López-González MA. Adding chloroquine to conventional treatment for glioblastoma multiforme: a randomized, double-blind, placebo-controlled trial. Ann Intern Med. 2006;144(5):337- 43. DOI: 10.7326/0003-4819-144-5-200603070-00008 PMID: 16520474 32. Rojas-Puentes LL, Gonzalez-Pinedo M, Crismatt A, Ortega-Gomez A, Gamboa-Vignolle C, Nuñez-Gomez R, et al. Phase II randomized, double-blind, placebo-controlled study of whole-brain irradiation with concomitant chloroquine for brain metastases. Radiat Oncol. 2013;8(1):209. DOI: 10.1186/1748-717X-8-209 PMID: 24010771 33. Eldredge HB, Denittis A, Duhadaway JB, Chernick M, Metz R, Prendergast GC. Concurrent whole brain radiotherapy and short-course chloroquine in patients with brain metastases: a pilot trial. J Radiat Oncol. 2013;2(3):315-21. DOI: 10.1007/s13566-013-0111-x PMID: 24187608 34. Kimura T, Takabatake Y, Takahashi A, Isaka Y. Chloroquine in cancer therapy: a double-edged sword of autophagy. Cancer Res. 2013;73(1):3- 7. DOI: 10.1158/0008-5472.CAN-12-2464 PMID: 23288916 35. Ben-Zvi I, Kivity S, Langevitz P, Shoenfeld Y. Hydroxychloroquine: from malaria to autoimmunity. Clin Rev Allergy Immunol. 2012;42(2):145-53. DOI: 10.1007/s12016-010-8243-x PMID: 21221847 36. Townsend KN, Hughson LR, Schlie K, Poon VI, Westerback A, Lum JJ. Autophagy inhibition in cancer therapy: metabolic considerations for antitumor immunity. 2012;249(1):176-94. DOI: 10.1111/j.1600- 065X.2012.01141.x PMID: 22889222 37. Loehberg CR, Strissel PL, Dittrich R, Strick R, Dittmer J, Dittmer A, et al. Akt and p53 are potential mediators of reduced mammary tumor growth by cloroquine and the mTOR inhibitor RAD001. Biochem Pharmacol. 2012;83(4):480-8. DOI: 10.1016/j.bcp.2011.11.022 PMID: 22142888 38. Maes H, Kuchnio A, Peric A, Moens S, Nys K, De Bock K, et al. Tumor vessel normalization by chloroquine independent of autophagy. Cancer Cell. 2014;26(2):190-206. DOI: 10.1016/j.ccr.2014.06.025 PMID: 25117709 224 CYTOLOGY, ONCOLOGY, CANCEROLOGY Zdrav Vestn | May – June 2022 | Volume 91 | https://doi.org/10.6016/ZdravVestn.3188 39. Antonelli M, Strappazzon F, Arisi I, Brandi R, D’Onofrio M, Sambucci M, et al. ATM kinase sustains breast cancer stem-like cells by promoting ATG4C expression and autophagy. Oncotarget. 2017;8(13):21692-709. DOI: 10.18632/oncotarget.15537 PMID: 28423511 40. Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H, Huang TH, et al. Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells. 2014;32(9):2309-23. DOI: 10.1002/stem.1746 PMID: 24809620 41. Sakamaki J, Wilkinson S, Hahn M, Tasdemir N, O’Prey J, Clark W, et al. Brodomain protein BRD4 is a transciptional repressor of autophagy and lysosomal function. Mol Cell. 2017;66(4):P517-32. DOI: 10.1016/j. molcel.2017.04.027 PMID: 28525743 42. Jang JE, Eom JI, Jeung HK, Cheong JW, Lee JY, Kim JS, et al. AMPK-ULK1- Mediated Autophagy Confers Resistance to BET Inhibitor JQ1 in Acute Myeloid Leukemia Stem Cells. Clin Cancer Res. 2017;23(11):2781-94. DOI: 10.1158/1078-0432.CCR-16-1903 PMID: 27864418 43. Hui B, Shi YH, Ding ZB, Zhou J, Gu CY, Peng YF, et al. Proteasome inhibitor interacts synergistically with autophagy inhibitor to suppress proliferation and induce apoptosis in hepatocellular carcinoma. Cancer. 2012;118(22):5560-71. DOI: 10.1002/cncr.27586 PMID: 22517429 44. Liang DH, Choi DS, Ensor JE, Kaipparettu BA, Bass BL, Chang JC. The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair. Cancer Lett. 2016;376(2):249-58. DOI: 10.1016/j. canlet.2016.04.002 PMID: 27060208 45. 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(1):628. DOI: 10.1186/s12879-014-0628-7 PMID: 25427651 46. Kresken M, Körber-Irrgang B. In vitro activity of nitroxoline against Escherichia coli urine isolates from outpatient departments in Germany. Antimicrob Agents Chemother. 2014;58(11):7019-20. DOI: 10.1128/ AAC.03946-14 PMID: 25182654 47. Cherdtrakulkiat R, Boonpangrak S, Sinthupoom N, Prachayasittikul S, Ruchirawat S, Prachayasittikul V. Derivatives (halogen, nitro and amino) of 8-hydroxyquinoline with highly potent antimicrobial and antioxidant activities. Biochem Biophys Rep. 2016;6:135-41. DOI: 10.1016/j. bbrep.2016.03.014 PMID: 29214226 48. Pelletier C, Prognon P, Bourlioux P. Roles of divalent cations and pH in mechanism of action of nitroxoline against Escherichia coli strains. Antimicrob Agents Chemother. 1995;39(3):707-13. DOI: 10.1128/ AAC.39.3.707 PMID: 7793877 49. Oviedo P, Quiroga M, Pegels E, Husulak E, Vergara M. Effects of subinhibitory concentrations of ciprofloxacin on enterotoxigenic Escherichia coli virulence factors. J Chemother. 2000;12(6):487-90. DOI: 10.1179/joc.2000.12.6.487 PMID: 11154030 50. Fraser RS, Creanor J. Rapid and selective inhibition of RNA synthesis in yeast by 8-hydroxyquinoline. Eur J Biochem. 1974;46(1):67-73. DOI: 10.1111/j.1432-1033.1974.tb03597.x PMID: 4854024 51. Sobke A, Klinger M, Hermann B, Sachse S, Nietzsche S, Makarewicz O, et al. The urinary antibiotic 5-nitro-8-hydroxyquinoline (Nitroxoline) reduces the formation and induces the dispersal of Pseudomonas aeruginosa biofilms by chelation of iron and zinc. Antimicrob Agents Chemother. 2012;56(11):6021-5. DOI: 10.1128/AAC.01484-12 PMID: 22926564 52. Mrhar A, Kopitar Z, Kozjek F, Presl V, Karba R. Clinical pharmacokinetics of nitroxoline. Int J Clin Pharmacol Biopharm. 1979;17(12):476-81. PMID: 118941 53. Wagenlehner FM, Münch F, Pilatz A, Bärmann B, Weidner W, Wagenlehner CM, 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(2):713-21. DOI: 10.1128/AAC.02147-13 PMID: 24217699 54. Wijma RA, Huttner A, Koch BC, Mouton JW, Muller AE. Review of the pharmacokinetic properties of nitrofurantoin and nitroxoline. J Antimicrob Chemother. 2018;73(11):2916-26. DOI: 10.1093/jac/dky255 PMID: 30184207 55. Shim JS, Matsui Y, Bhat S, Nacev BA, Xu J, Bhang HE, et al. Effect of nitroxoline on angiogenesis and growth of human bladder cancer. J Natl Cancer Inst. 2010;102(24):1855-73. [cited 2013 Nov 18]. DOI: 10.1093/jnci/ djq457 PMID: 21088277 56. Xu N, Lin W, Sun J, Sadahira T, Xu A, Watanabe M, et al. Nitroxoline inhibits bladder cancer progression by reversing EMT process and enhancing anti-tumor immunity. J Cancer. 2020;11(22):6633-41. DOI: 10.7150/ jca.47025 PMID: 33046984 57. Mirković B, Renko M, Turk S, Sosič I, Jevnikar Z, Obermajer N, et al. Novel mechanism of cathepsin B inhibition by antibiotic nitroxoline and related compounds. ChemMedChem. 2011;6(8):1351-6. DOI: 10.1002/ cmdc.201100098 PMID: 21598397 58. Mirković B, Markelc B, Butinar M, Mitrović A, Sosič I, Gobec S, et al. Nitroxoline impairs tumor progression in vitro and in vivo by regulating cathepsin B activity. Oncotarget. 2015;6(22):19027-42. 59. Vasiljeva O, Turk B. Dual contrasting roles of cysteine cathepsins in cancer progression: apoptosis versus tumour invasion. Biochimie. 2008;90(2):380-6. DOI: 10.1016/j.biochi.2007.10.004 PMID: 17991442 60. Gocheva V, Zeng W, Ke D, Klimstra D, Reinheckel T, Peters C, et al. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev. 2006;20(5):543-56. DOI: 10.1101/gad.1407406 PMID: 16481467 61. Chan-On W, Huyen NT, Songtawee N, Suwanjang W, Prachayasittikul S, Prachayasittikul V. Quinoline-based clioquinol and nitroxoline exhibit anticancer activity inducing FoxM1 inhibition in cholangiocarcinoma cells. Drug Des Devel Ther. 2015;9:2033. PMID: 25897210 62. Mitrović A, Kos J. Nitroxoline: repurposing its antimicrobial to antitumor application. Acta Biochim Pol. 2019;66(4):521-31. PMID: 31834689 63. Veschi S, De Lellis L, Florio R, Lanuti P, Massucci A, Tinari N, et al. Effects of repurposed drug candidates nitroxoline and nelfinavir as single agents or in combination with erlotinib in pancreatic cancer cells. J Exp Clin Cancer Res. 2018;37(1):236. DOI: 10.1186/s13046-018-0904-2 PMID: 30241558 64. Sosič I, Mirković B, Arenz K, Štefane B, Kos J, Gobec S. Development of new cathepsin B inhibitors: combining bioisosteric replacements and structure-based design to explore the structure-activity relationships of nitroxoline derivatives. J Med Chem. 2013;56(2):521-33. DOI: 10.1021/ jm301544x PMID: 23252745 65. Sosič I, Mitrović A, Ćurić H, Knez D, Brodnik Žugelj H, Štefane B, et al. Cathepsin B inhibitors: further exploration of the nitroxoline core. Bioorg Med Chem Lett. 2018;28(7):1239-47. DOI: 10.1016/j.bmcl.2018.02.042 PMID: 29503024 66. Mitrović A, Sosič I, Kos Š, Tratar UL, Breznik B, Kranjc S, et al. Addition of 2-(ethylamino)acetonitrile group to nitroxoline results in significantly improved anti-tumor activity in vitro and in vivo. Oncotarget. 2017;8(35):59136-47. DOI: 10.18632/oncotarget.19296 PMID: 28938624 67. Bhat S, Shim JS, Zhang F, Chong CR, Liu JO. Substituted oxines inhibit endothelial cell proliferation and angiogenesis. Org Biomol Chem. 2012;10(15):2979-92. DOI: 10.1039/c2ob06978d PMID: 22391578 68. Chen W, Zhang H, Chen Z, Jiang H, Liao L, Fan S, et al. Development and evaluation of a novel series of Nitroxoline-derived BET inhibitors with antitumor activity in renal cell carcinoma. Oncogenesis. 2018;7(11):83. DOI: 10.1038/s41389-018-0093-z PMID: 30385738 69. Xing J, Zhang R, Jiang X, Hu T, Wang X, Qiao G, et al. Rational design of 5-((1H-imidazol-1-yl)methyl)quinolin-8-ol derivatives as novel bromodomain-containing protein 4 inhibitors. Eur J Med Chem. 2019;163:281-94. DOI: 10.1016/j.ejmech.2018.11.018 PMID: 30529546 70. Mitrović A, Kljun J, Sosič I, Uršič M, Meden A, Gobec S, et al. Organoruthenated Nitroxoline Derivatives Impair Tumor Cell Invasion through Inhibition of Cathepsin B Activity. Inorg Chem. 2019;58(18):12334- 47. DOI: 10.1021/acs.inorgchem.9b01882 PMID: 31464130 225 REVIEW ARTICLE Possible use of antimicrobial agents in cancer therapy 71. Hu D, Xu H, Xiao B, Li D, Zhou Z, Liu X, et al. Albumin-Stabilized Metal- Organic Nanoparticles for Effective Delivery of Metal Complex Anticancer Drugs. ACS Appl Mater Interfaces. 2018;10(41):34974-82. DOI: 10.1021/ acsami.8b12812 PMID: 30238746 72. Oliphant CM, Green GM. Quinolones: a comprehensive review. Am Fam Physician. 2002;65(3):455-64. PMID: 11858629 73. Dalhoff A. Immunomodulatory activities of fluoroquinolones. Infection. 2005;33(S2):55-70. DOI: 10.1007/s15010-005-8209-8 PMID: 16518713 74. Herold C, Ocker M, Ganslmayer M, Gerauer H, Hahn EG, Schuppan D. Ciprofloxacin induces apoptosis and inhibits proliferation of human colorectal carcinoma cells. Br J Cancer. 2002;86(3):443-8. DOI: 10.1038/ sj.bjc.6600079 PMID: 11875713 75. Aranha O, Grignon R, Fernandes N, McDonnell TJ, Wood DP, Sarkar FH. Suppression of human prostate cancer cell growth by ciprofloxacin is associated with cell cycle arrest and apoptosis. Int J Oncol. 2003;22(4):787-94. DOI: 10.3892/ijo.22.4.787 PMID: 12632069 76. Phiboonchaiyanan PP, Kiratipaiboon C, Chanvorachote P. Ciprofloxacin mediates cancer stem cell phenotypes in lung cancer cells through caveolin-1-dependent mechanism. Chem Biol Interact. 2016;250:1-11. DOI: 10.1016/j.cbi.2016.03.005 PMID: 26947806 77. Chen TC, Hsu YL, Tsai YC, Chang YW, Kuo PL, Chen YH. Gemifloxacin inhibits migration and invasion and induces mesenchymal-epithelial transition in human breast adenocarcinoma cells. J Mol Med (Berl). 2014;92(1):53-64. DOI: 10.1007/s00109-013-1083-4 PMID: 24005829 78. Kan JY, Hsu YL, Chen YH, Chen TC, Wang JY, Kuo PL. Gemifloxacin, a fluoroquinolone antimicrobial drug, inhibits migration and invasion of human colon cancer cells. BioMed Res Int. 2013;2013:159786. DOI: 10.1155/2013/159786 PMID: 24386633 79. Gong JH, Liu XJ, Shang BY, Chen SZ, Zhen YS. HERG K+ channel related chemosensitivity to sparfloxacin in colon cancer cells. Oncol Rep. 2010;23(6):1747-56. PMID: 20428834 80. Shan G, Li Y, Zhang J, Li W, Szulwach KE, Duan R, et al. A small molecule enhances RNA interference and promotes microRNA processing. Nat Biotechnol. 2008;26(8):933-40. DOI: 10.1038/nbt.1481 PMID: 18641635 81. Valianatos G, Valcikova B, Growkova K, Verlande A, Mlcochova J, Radova L, et al. A small molecule drug promoting miRNA processing induces alternative splicing of MdmX transcript and rescues p53 activity in human cancer cells overexpressing MdmX protein. PLoS One. 2017. ;12(10)DOI: 10.1371/journal.pone.0185801 PMID: 28973015 82. Katsarou ME, Efthimiadou EK, Psomas G, Karaliota A, Vourloumis D. Novel copper(II) complex of N-propyl-norfloxacin and 1,10-phenanthroline with enhanced antileukemic and DNA nuclease activities. J Med Chem. 2008;51(3):470-8. DOI: 10.1021/jm7013259 PMID: 18205294 83. Hu G, Yang Y, Yi L, Wang G, Duan N, Wen X, et al. Design, synthesis and antitumor activity of C3/C3 bis-fluoroquonolones cross-linked with [1,2,4]triazolo[3,4-b] [1,3,4]thiadiazole. Acta Pharm Sin B. 2011;1(3):172- 7. DOI: 10.1016/j.apsb.2011.07.001