Next Article in Journal
Facile Fabrication of 1-Methylimidazole/Cu Nanozyme with Enhanced Laccase Activity for Fast Degradation and Sensitive Detection of Phenol Compounds
Next Article in Special Issue
Copper(II) Complex Containing 4-Fluorophenoxyacetic Acid Hydrazide and 1,10-Phenanthroline: A Prostate Cancer Cell-Selective and Low-Toxic Copper(II) Compound
Previous Article in Journal
Literature Mining of Disease Associated Noncoding RNA in the Omics Era
Previous Article in Special Issue
Porphyrins and Metalloporphyrins Combined with N-Heterocyclic Carbene (NHC) Gold(I) Complexes for Photodynamic Therapy Application: What Is the Weight of the Heavy Atom Effect?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evidence of Metallic and Polyether Ionophores as Potent Therapeutic Drug Candidate in Cancer Management

1
Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida 201306, India
2
Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
3
Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
4
Department of Electrical & Computer Engineering, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia
5
Center of Excellence in Environmental Studies, King Abdulaziz University, Jeddah 21589, Saudi Arabia
6
Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Sendai 980-8577, Japan
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(15), 4708; https://doi.org/10.3390/molecules27154708
Submission received: 27 June 2022 / Revised: 18 July 2022 / Accepted: 20 July 2022 / Published: 23 July 2022

Abstract

:
Cancer remains one of the most crucial human malignancies with a higher mortality rate globally, and is predicted to escalate soon. Dysregulated ion homeostasis in cancerous cells prompted the researchers to investigate further ion homeostasis impeding agents as potent anticancerous agents. Reutilization of FDA-approved non-cancerous drugs has emerged as a practical approach to developing potent, cost-effective drugs for cancer treatment. Across the globe, most nations are incapable of fulfilling the medical demands of cancer patients due to costlier cancerous drugs. Therefore, we have inclined our review towards emphasizing recent advancements in cancer therapies involving ionophores utilization in exploring potent anticancer drugs. Numerous research reports have established the significant anticancerous potential of ionophores in several pre-clinical reports via modulating aberrant cell signaling pathways and enhancing antitumor immunity in immune cells. This review has mainly summarized the most significant ion homeostasis impeding agents, including copper, zinc, calcium, and polyether, that presented remarkable potential in cancer therapeutics via enhanced antitumor immunity and apoptosis induction. Altogether, this study could provide a robust future perspective for developing cost-effective anticancerous drugs rapidly and cost-effectively, thereby combating the limitations of currently available drugs used in cancer treatment.

1. Introduction

Enormous evidence strongly indicates the significance of altered ion transport in cancer progression [1]. Cancerous cells control their growth and proliferation by maintaining their ion pumps, ion homeostasis, and ion channels which are too crucial for their growth and survival [2]. Controlled regulation of the maintenance of ions in the extracellular matrix and inside the cells is vital for proper cell shape, maintained membrane potential, and normal cell signaling pathways. The structural organization of the plasma membrane leads to the impermeability of ions, macromolecules (e.g., RNA), and several other hydrophilic molecules (e.g., glucose) across the cellular membrane [3]. However, cells overrule these transport phenomena by developing several mechanisms for active transport and facilitated diffusion of molecules and ions across membranes. Facilitated transport constitutes ion diffusion towards a protein-mediated concentration gradient that leads to the formation of water-filled ion channels across the cell membrane. The gating of ion channels is controlled (opened or closed) depending on cellular requirements. Commonly reported gated ion channels include voltage-gated, ligand-gated, light-gated, and mechanically gated [4,5,6].
During normal cellular functioning, ions (or molecules) are transported using numerous active transporter proteins (H+/K+ ATPase, ABC transporters Ca2+ ATPase, ATP. Na+/K+ ATPase) against the concentration gradient [7,8,9]. Aberrant functioning (expression) of these ion channels and pumps in cancerous cells helps them regulate distinctive ion homeostasis that further helps them in growth and proliferation [10,11]. Ionophores (calcium, copper, zinc, and polyether) have widely been utilized in cancer treatment as chemo-sensitizers for maintaining the ion balance in cancerous cells [12,13,14]. Therefore, we have accumulated research evidence explaining how cancer cells utilize ion homeostasis for their growth and providing a potential cancer therapeutic approach. Further sections will uncover the anti-cancerous potential of numerous potent metal ionophores such as copper, calcium, and zinc.

2. Repurposing of Copper Ionophores in Cancer

Bioactive compounds, including flavones (O, O donor ligands), have been classified as copper ionophores with significant anticancerous potential. Numerous copper (Cu) ionophores have displayed a wider range of anticancerous potential either by ROS-mediated cytotoxicity or targeting specific signaling pathways [13,14,15,16]. Copper is preferentially transported to cuproenzymes in mitochondria, such as the antioxidant enzyme superoxide dismutase 1 and the respiratory chain complex IV (COX) (SOD1). The cuproenzyme COX participates in the mitochondrial respiratory chain by catalyzing the reduction of molecular oxygen to water. Copper plays a significant role in COX-mediated ATP synthesis, indicating that this bio-metal center is essential for maintaining essential functions. Important copper/zinc-containing antioxidant enzyme SOD1 (superoxide dismutase 1) is primarily found in the cytosol and in the intermembrane space of mitochondria. Furthermore, intracellular copper alters the mitochondrial function or triggers metabolic reprogramming to affect cancer cell proliferation or differentiation.
Copper (Cu) ionophores can be grouped into some significant classes, such as thiosemicarbazones (C10H12N4OS) (TSCs) and dithiocarbamates (DTCs), hydroxyquinolines (C9H7NO4S) (HQs), and hydroxyflavones (HFs) [17,18]. It is equally important to study the copper complexes’ physicochemical potential to determine the copper ionophores’ efficacy. Pyrrolidine dithiocarbamate (C5H9NS2) (PDTC) was the first identified Cu ionophores that showed its toxic efficacies via transport of redox-active copper into the cell that further inhibited cell proliferation, ubiquitin-proteasome/nuclear factor-jB mediated apoptosis induction in prostate and breast cancerous cells [19,20]. DTCs have exhibited significant metal uptake and apoptosis induction in numerous cancer cells. Disulfiram (C10H20N2S4) (DSF) is highly effective against various carcinomas such as prostate and breast carcinoma via targeting stem-like cancer cells and potentiating the cytotoxic efficacies of a broader range of anticancer drugs. However, phase II clinical trials of DSF in prostate cancer patients failed to produce significant benefits. Thus, DSF could only affect tumor growth of castrate-resistant and hormone-sensitive prostate cancer (xenografts) when co-administered with copper [21].
Numerous research studies reported ROS augmentation in cells treated with Cu and DSF, mainly due to the redox cycling of CuDTC (copper dithiocarbamates) complexes [22,23]. Cu-DSF (copper disulfiram) inhibited NF-kB (Nuclear factor-kappa B) activation and down-regulated XIAP (apoptotic protein and chemoresistance factor), thereby leading to pro-apoptotic response activation [24]. Cu-DSF drug not only triggered ROS but simultaneously inhibited NF-jB up-regulation resulting in potent improvements in ROS-targeted chemotherapeutic approaches [25]. A possible mechanism behind these improvements could be the involvement of copper ion since Cu-XIAP (copper X-linked inhibitor of apoptosis protein) complex formation induce apoptosis induction by inhibiting the anti-apoptotic potential of XIAP [26]. Cu-DSF also inhibited ALDH1 (Aldehyde Dehydrogenase 1), a potent anti-cancer target that can potentially sensitize cancer stem cells to traditional chemotherapeutic cancer drugs. Other targets of Cu-DSF have also been unrevealed, such as the p-97-NPL4-UFDI protein complex that mainly regulates proteasomes upstream, and its inhibition might inhibit proteasomes leading to ubiquitinated proteins accumulation [27].
Co-administration of Cu and DSF is selectively toxic to several malignant cells compared to healthy prostate cells. This property could be attributed to the ability of prostate cancer cells to accumulate Cu and the oxidative environment inside the cancerous cells, making them susceptible to ROS (reactive oxygen species). Further research displayed that Cu ionophores such as DSF could selectively target prostate cancer cells by releasing intra-cellularly Cu leading to ROS formation in TRAMP (transgenic adenocarcinoma of the mouse prostate) adenocarcinoma cells (TRAMP-C1). TRAMP-C1 cells have a lower ability to protect from oxidative stress [28]. Cu-DSF treatment reduced GSH levels in prostate cancer cells and increased intracellular GSH (reduced form of glutathione) in normal prostate epithelial cells [29]. Co-administration of Cu and 2, 20-dithiodipyridine (DPy) is highly cytotoxic against various cancer cells, including HEPG2 (human liver cancer cell line), SMMC-7721 (human hepatoma cell line), HeLa (cervical cancer cells), and A549 (human lung cancer cells) cancer cells via enhancing intracellular Cu levels in a time-dependent manner in comparison to their effects when either of them is administered alone [30]. Moreover, their synergistic efficacies (DPy and Cu) have not been reported on normal cells such as BEAS-2B (epithelial cell line), L02 (cellosaurus cell line), and HEK 293 (human embryonic kidney) T cells [31].
After that, the research shifted towards developing novel bis (thiosemicarbazone) derivatives with reduced toxicity and similar biological activity. Co-administration of two thiosemicarbazone derivatives (ATSM and GTSM) along with copper, namely Cu-GTSM and Cu-ATSM (Cu prototypes), have various cell metabolism and reduction potential and have selectively targeted prostate cancer cells [32]. Co-administration with Cu makes them more active than the corresponding ligand, and an increase in intracellular Cu concentration is associated with their activity. Cu-ATSM is effective at higher doses than Cu-GTSM against different cancer cells. Cu-GTSM also mediated its anti-proliferative potential via increased ROS generation, cathepsin release, lysosomal membrane permeabilization, and apoptotic cascades [33] (Table 1).
Cu ionophores represent a promising therapeutic approach for managing metal dyshomeostasis diseases [42,43]. Long-term usage of ionophores might perturb the usual physiological roles of essential metalloproteins by affecting the homeostasis of biometals. Increased copper levels in cancerous tissues and ionophore administration make cancerous cells more susceptible to ionophore-copper toxicity than normal cells. However, a higher therapeutic selectivity (normal versus cancerous cells) is needed to elucidate a potent drug. Conjugating ionophores with targeting moieties might reduce metal-binding agents’ toxicities and further increase their efficacies [44]. Protecting the copper-binding site might also avoid unwanted systemic chelation. These targets can be easily achieved by specific activation of metal ionophores at their site of action.
Proinophores (site-specific activation of prodrugs) can be achieved by utilizing numerous specific intracellular enzymes, which could further emphasize the urge to target elective enzymes overexpressed in cancerous cells. These enzymes must fully hydrolyze proionophores in cancerous cells to exhibit maximum anti-cancerous potential [45]. The glucoconjugation strategy has been widely used to develop proionophores since they possess numerous benefits, including water solubility, limiting side effects, and enhanced targeting [46]. Galactose, glucose, and moieties have been used with hydroxyquinolines (HQ) molecules to obtain proionophores [47].

3. Calcium, Zinc and Potassium Ionophores in Cancer

Ca2+ ion contributes significantly to numerous cell signaling pathways that further control a more comprehensive range of cellular processes via interacting with various signaling cascades. Calcium ions are essential in cytosolic signaling as second messengers, and thus cytosolic Ca2+ (Ca2+i) concentration remains a significant regulator of vital cellular phenomena, including cell proliferation and apoptosis [48]. Thus, intracellular Ca2+ concentration should always be adequately maintained by calcium-permeable channels such as store-operated channels (SOCs), transient receptor potential (TRP) channels, mitochondrial calcium uniporter (MCU), and others for normal cell functioning. Ca2+/calcineurin and Ca2+/calmodulin have emerged as significant checkpoints during cell cycle progression. Ca2+-dependent metastasis processes include cell deformation, migration, invasion, and adhesion [49,50]. TRPM7 (transient receptor potential ion channel subfamily M, member 7) channels have been involved in the formation of transient and local calcium domains (calcium flickers) at lamellipodia which guides migration direction [51,52]. Ca2+ transport-mediated growth and metastasis are reported in prostate, lung, and breast carcinomas [52]. It has also been observed that apoptotic cells augment intrinsic Ca2+ either by releasing calcium from stressed ER or by continual Ca2+ influx through activated channels. Cancerous cells display higher apoptotic resistance by calcium influx inhibition either via down-regulating channels or adjusting chronic-reduced ER Ca2+ [53].

3.1. Zinc Ionophores in Cancer

Research has displayed a strong association between zinc and cancer development. Different malignancies are known to develop and progress in response to zinc imbalance. Evidence also suggests that disruption of FCZ homeostasis occurs early in cancer development. Reduced zinc levels are generally detected in neoplastic tissue, and zinc transporter expression may also play a role in carcinogenesis. It is interesting to note that prostate cancer has been treated with higher levels of cellular zinc or zinc plus cadmium. Remember that the 2+ oxidation state of cadmium is stabilized by the presence of a core of filled subshells. Zinc ionophores have been used to increase zinc transport across plasma membranes [Zn2+]. Notably, compared to more traditional medicines, zinc-based cancer therapy methods seem to have substantially lower levels of cytotoxicity. Zinc cations interfere with anaphase-A chromosome movement and mitosis through negative shielding charges bounded at microtubule and centrosomes. Cell responses to Zn2+ are highly significant for chemotherapeutics. Zinc dysregulation has been implicated in the development and progression of various carcinomas, including deregulation of free cytosolic zinc concentration (FCZ) early homeostasis in cancer development. Zinc transporter expression has also been reported in carcinogenesis, while reduced zinc levels were reported in neoplastic tissue. Increased cellular zinc or zinc+cadmium has shown significant potential in prostate cancer treatment with minimal cytotoxicity. Zn2+ concentration is augmented by utilizing zinc ionophores to facilitate zinc transport across the plasma membrane [54]. For instance, Clioquinol (zinc ionophore) exhibited potential anticancer activity. Chloroquine (antimalarial agent), along with zinc, has also shown significant efficacy in clinical trials of human ovarian cancer cells (A2780) via inducing apoptosis [55].

3.2. Potassium Ionophores in Cancer

Programmed cell death may be affected by changes in the transmembrane gradients of some physiological ions. According to recent studies, apoptosis may be promoted or inhibited by intracellular calcium increases depending on their level, timing, and location. In contrast, intracellular potassium loss is thought to promote apoptosis. Strong evidence currently suggests that excessive potassium (K+) efflux and intracellular K+ depletion are critical early steps in apoptosis, in contrast to the ionic process of necrosis, which involves Ca2+ influx and intracellular Ca2+ buildup. Apoptotic effectors are suppressed by intracellular K+ concentration at physiological levels. Activating crucial steps in the apoptotic cascade, such as caspase cleavage, cytochrome c release, and endonuclease activation, a significant loss of cellular K+, which is probably a common occurrence in the apoptosis of many cell types, may act as a disaster signal that permits the execution of the suicide program. K+ homeostasis disturbance that promotes apoptosis can be caused by excessively active K+ channels or ionotropic glutamate receptor channels and is most often accompanied by decreased K+ uptake because of Na+, K+-ATPase failure. Four ionophores were created for potassium: hemispheres, mono- and biscrown ethers (like potassium ionophore III), and antibiotics. Due to its excellent selectivity and widespread use in clinical analyzers, valinomycin is the most used K+ ionophore. Because they are inexpensive, mono crown ether sensors frequently exhibit a modest level of selectivity enhancement. Biscrown ethers, which are also capable of forming intramolecular sandwich complexes and interacting with the internal cavity of the crown ether ring, produce noticeably superior results. From this review, it can be concluded that maintaining zinc, calcium, copper, and potassium homeostasis is essential for maintaining human health and immunity.

4. Natural Polyether Ionophores Antibiotics as Potent Anticancer Agents

Natural polyether ionophores are potent antibiotics that belong to the families of naturally occurring ionophores. It has been evident that ionophores refer to the binding of a molecule to a metal ion that facilitates its transportation via the cell membrane. These chemical and physiological properties of polyether ionophores made it a valuable tool for studying the mechanisms associated with cation transport. Polyether ionophores (squalene derivatives) exhibit a wider range of higher biological activities. Polyether ionophores are highly significant and sizable classes of naturally occurring chemicals. In recent years, there has been an increase in interest in this kind of chemical. Over 120 known naturally occurring ionophores have been identified. Controlling coccidiosis is the primary commercial use of ionophores. They are also applied to ruminants to encourage growth. These substances increase production efficiency by precisely targeting the ruminal bacterial population. Ionophores were the antimicrobials most frequently employed in beef cattle production in 2003. The ionophores with the highest combined annual sales were lasalocid (C34H54O8) (Avatec, Bovatec, Australia), monensin (C36H62O11) (Coban, Rumensin, and Coxidin, United States), and salinomycin (C42H70O11) (Bio-cox, Sacox), narasin (C43H72O11) (Monteban, Maxiban), maduramycin (C47H83NO17) (Cygro, United States), and laidlomycin propionate (C40H66O13) (Cattlyst, United States). Ion-selective electrodes can be made using ionophores as well (Figure 1).
Phycotoxins (marine polyether ionophores) are primarily produced by phytoplankton, fungi, diatoms, cyanobacteria, dinoflagellates, bacteria, and a few marine invertebrates [56,57]. Anticancer antibiotics can inhibit the growth progression of cancer cells in every stage of the cell proliferation cycle (at G0, S, M/G2 phase) by inducing cell cycle arrest, such as cyclin non-specific drugs. In contrast, other antibiotics could induce apoptosis by targeting specific apoptotic genes such as Bcl-2/Bax, caspase-3/8/9, and p53 in cancer patients. These antibiotics further could be exploited as anti-metastasis or EMT regulatory agents for the inhibition of metastasis of cancer cells, such as ciprofloxacin (C17H18FN3O3) (pro-apoptosis role), salinomycin (growth inhibition), and nigericin [58,59,60]. Numerous research has speculated that cancer patients seem more sensitive to these emerging anticancer antibiotics that have not been previously exposed. Anticancer antibiotics have been increasingly essential, from the first anticancer antibiotics such as doxorubicin (C27H29NO11), epirubicin (C27H29NO11), and mitomycin (C15H18N4O5) to the anticancer antibiotics adriamycin, salinomycin, and fluoroquinolones discovered in recent years. Thus, we have summarized two major antibiotics that have been recently repurposed for cancer treatment, such as nigericin and salinomycin, in subsequent sections.

4.1. Nigericin as Potent Therapeutic Candidate for Cancer Management

Nigericin (antibiotic) derived from Streptomyces peucetius has been recognized as a potent polyether ionophore used mainly in veterinary medicine. Nigericin has exhibited promising anticancerous potential in several carcinomas, including prostate cancer, colorectal cancer, ovarian cancer, bladder carcinoma, leukemia, pancreatic cancer, and breast carcinoma. Figure 2 displays the efficacy of nigericin against anticancerous drugs via reducing pH and increasing the toxicity of anticancerous drugs. Extracellular pH of tumor is generally lower (about 0.5 pH units) than normal tissues, and cells might survive in these acidic conditions due to the antiports in their membrane exchanges of HCO3− for Cl and Na+ for H+ that helps in regulating the intracellular pH. Later, DNA synthesis inhibition was reported in nigericin-treated cancer cells without significant reduction in MMP or ATP concentration [61,62]. Further research reported nigericin-induced alterations in intracellular ion concentrations in neurons, synaptosomes, and C6 glioma cells.
Recent research reported loss of K+ and intracellular acidification, increased lactate production, oxygen consumption, and reduced cellular energy level in nigericin-treated cancer cells [63]. Nigericin (K+ ionophore) exhibited significant inhibitory potential against several essential target proteins of Wnt signaling pathways such as Wnt5a/b, LRP6, and β-catenin. Research further reported significant nigericin-induced cell growth inhibition during prolonged serum starvation, normal culture conditions, and lung tumorspheres [64,65]. Wang et al. reported the dose-dependent nigericin induced inhibited invasion and migration of epithelial ovarian cells via suppressing the EMT transition.
In 1998, Zanke et al. reported nigericin-mediated intracellular acidification leading to cell death induction by activating JNK or stress-activated protein kinases (SAPK) signaling pathways and Bax expression in human MGH-U1 (bladder cancer) cells [66]. Later, nigericin showed remarkable inhibitory potential in colorectal cancers by directly targeting the critical β-catenin destruction complex. Nigericin was also identified as a potent gp170 inhibitor (cancer stem cells inhibitor) in MDR cancer cells and was also reported to kill selective cancer stem cells and overcome multidrug resistance in malignant tumors [67]. In nasopharyngeal cancer, nigericin was also reported to target cancer stem cells and sensitize them to cisplatin drugs in both in vivo and in vitro studies [68]. Although nigericin exhibited promising anticancerous efficacies in several malignancies, its adverse effects cannot be ignored. Thus, it is imperative to develop other drug administration systems for increased bioavailability, avoidance of adverse effects, and improved therapeutic potential.

4.2. Salinomycin as Potent Therapeutic Candidate for Cancer Management

Salinomycin is a natural polyether antibiotic isolated from the Streptomyces albus strain. Gupta et al. (2009) reported salinomycin as a potent anticancer drug for the first time while screening approximately 16,000 compounds for their anti-cancerous potential against cancer stem cells [69,70]. They reported salinomycin as a more effective anticancer drug than paclitaxel drug. Later, other research studies have also projected its selectivity in targeting cancer stem cells in various cancer cells. Functionally, SAL is one of the membrane ionophores that exhibits a strong affinity for alkali cations, particularly for K. Mounting evidence implicates SAL’s ion transport capabilities as the cause of its phenotypic effects. Apoptosis in human lymphoma cells was previously thought to be induced by a drop in intracellular K concentration. Potassium channels of the mitochondrial and cytoplasmic membranes, which are overexpressed in many human cancer cells, play crucial roles in the regulation of tumorigenesis, tumor cell proliferation, cell cycle progression, and apoptosis and may represent novel and promising molecular targets for cancer therapy.
Salinomycin exhibited these anti-cancerous efficacies via targeting multiple cell signaling pathways (Figure 3). It was shown that SAL treatment inhibits the Wnt signaling pathway, which is implicated in cancer and embryogenesis, by many mechanisms; other pathways that were blocked by SAL treatment include K-Ras (Kirsten rat sarcoma viral oncogene homolog) and regulation of Hedgehog signaling. It has been demonstrated that changes in genes associated with stemness-related pathways, such as Wnt, Notch, and Hedgehog, are crucial for the development of breast cancer [71,72,73,74]. Salinomycin treatment showed a significant increase in cell death in cancer stem cells and numerous cancer cells through apoptosis induction, enhanced oxidative stress, and autophagy inhibition [75,76]. Salinomycin modulated cancer metastasis via inhibition of cancer cell migration and invasion by targeting EMT (epithelial-mesenchymal transition), Hedgehog signaling and Wnt cell signaling pathways in several cancers, including breast cancer [77]. Salinomycin has shown significant potential in sensitizing several cancers and enhancing the efficacy of commonly used anti-cancer drugs, including doxorubicin, gemcitabine, trastuzumab, and tamoxifen, etc. [78,79,80]. Recent research by Zhang et al. reported increased sensitization to gemcitabine in salinomycin-treated pancreatic cancer by targeting cancer stem cells. Another study reported the sensitizing efficacy of salinomycin to resveratrol in breast cancer [81]. One of the major hindrances in cancer treatment is the resistance of cancerous cells to conventional therapies, and cells escape themselves from eradication. These cells become more aggressive and resistant, leading to cancer relapse.
Most current cancer therapeutic approaches are incompetent in achieving complete cancer elimination and result in cancer relapse after initial remission. Interestingly, salinomycin has also exhibited significant anticancerous potential against numerous multi-drug resistant cancers and developed a drug efflux mechanism by inhibiting several drug transporters such as p-glycoproteins [82]. One major drawback associated with the therapeutic application of salinomycin is toxicity and poor water solubility to normal cells. However, these limitations can get overruled by investigating targeted delivery approaches or synthesizing their more specific and less toxic analogs. Recent research has reported the successful utilization of nanomicelles, nanoparticles, nanotubes, and cell surface markers (CD133 and CD44) conjugated multilamellar liposomes for targeted and efficient delivery of salinomycin [83,84,85,86,87].
Other polyether ionophores, including calcimycin, alborixin, and monexin, have significant anticancerous potential. S100A4 was inhibited by calcimycin in a concentration- and time-dependent manner. Additionally, in cultured keloid fibroblasts (KFs), calcimycin reduced TGF-1-induced expression of collagen type I, fibronectin, smooth muscle actin, and cell viability. Additionally, calcimycin altered the production of Smad7 (SMAD Family Member 7), a TGF-/Smad target gene, and the phosphorylation of Smad2/3, a TGF-1/Smad1-induced gene. The discovery of S100A4 (hypoxia-inducible gene) in KFs was made possible by this study for the first time. S100A4 expression, KF migration, proliferative activity, and extracellular matrix (ECM) production are all inhibited by calcimycin. Together, these findings suggest that calcimycin may be a potential treatment for similar keloid or other fibrotic illnesses [88].
Alborixin exhibited maximum cytotoxic efficacy against human colon carcinoma (HCT-116) cells via elevating intracellular ROS levels accompanied by mitochondrial membrane potential (MMP) loss, reduced Bcl-2 (anti-apoptotic) protein expression level, augmented cleavage of PARP-1 and caspase-3, activated caspase-8, 9 and increased Bax (proapoptotic) protein expression levels [89]. Monesin displayed its proliferation suppression efficacy via inhibiting several growth factor-induced signaling pathways (EGFR). Monesin further reduces the expression level of cyclin D1, cyclin A, and CDK6 and stimulates mitochondria transmembrane potential against numerous human cancer cells [90]. Although oncology has made significant strides in the fight against cancer, people are still waiting for the creation of efficient treatments. Chemotherapy, whose mechanism of action is based on the prevention of cell division, is successful in treating some malignancies but ineffective in treating others. The hunt for novel physiologically active compounds is therefore of utmost significance. Natural substances have been widely employed to treat cancer over the past few decades, and a lot of research suggests that naturally occurring polyether antibiotic is a potential for a brand-new, highly potent anticancer medicine.

5. Conclusions

Ion homeostasis disruption is crucial for the survival and proliferation of cancer and comprises a potent target for chemotherapy. Our review has mainly focused on metallic and polyether ionophores’ therapeutic potential, emphasizing their anti-cancerous potential through targeting signaling pathways. Existing drugs, such as antibiotics, have been repositioned as antitumor agents in the recent decade, and the results of more clinical trials on their potency are awaited. Research in this area should target developing novel and effective therapeutics of known pharmaceuticals and exploring new drug delivery systems for managing cancer. Several ionophores, such as metal and polyether ionophores, have displayed potent anticancer potential in several preclinical cancer models, either alone or in combination with other potent anticancer drugs. These medications can be used in antitumor therapy at a lower cost, making treatment more affordable in developing countries. Altogether, the development of target-specific ionophores might provide a novel therapeutic approach for better management of carcinomas.

Author Contributions

Conceptualization, P.P. and F.K.; methodology, P.P. and F.K.; resources, P.P. and F.K.; writing—original draft preparation, P.P. and F.K.; visualization, P.P. and F.K.; supervision, P.P., F.K. and T.K.U.; project administration, P.P., F.K., A.F.A., H.A.Q. and M.O.; writing—review and editing, T.K.U., A.F.A., H.A.Q. and M.O.; funding acquisition, A.F.A., H.A.Q. and M.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the management of Noida Institute of Engineering and Technology, Greater Noida for its support. We are also highly grateful to the Dean of Scientific Research (DSR), King Abdulaziz University, Jeddah, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Biasiotta, A.; D’Arcangelo, D.; Passarelli, F.; Nicodemi, E.M.; Facchiano, A. Ion channels expression and function are strongly modified in solid tumors and vascular malformations. J. Transl. Med. 2016, 14, 285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Elingaard-Larsen, L.O.; Rolver, M.G.; Sørensen, E.E.; Pedersen, S.F. How Reciprocal Interactions Between the Tumor Microenvironment and Ion Transport Proteins Drive Cancer Progression. In From Malignant Transformation to Metastasis; Stock, C., Pardo, L.A., Eds.; Springer: Cham, Switzerland, 2020; Volume 182, pp. 1–38. [Google Scholar] [CrossRef]
  3. Choromańska, A.; Chwiłkowska, A.; Kulbacka, J.; Baczyńska, D.; Rembiałkowska, N.; Szewczyk, A.; Michel, O.; Gajewska-Naryniecka, A.; Przystupski, D.; Saczko, J. Modifi-cations of Plasma Membrane Organization in Cancer Cells for Targeted Therapy. Molecules 2021, 26, 1850. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, J.-Y.; Hou, J.-L. Controllable synthetic ion channels. Org. Chem. Front. 2018, 5, 1728–1736. [Google Scholar] [CrossRef]
  5. McGivern, J.G.; Ding, M. Ion channels and relevant drug screening approaches. SLAS DISCOV. Adv. Sci. Drug Discov. 2020, 25, 413–419. [Google Scholar] [CrossRef]
  6. Djamgoz, M.B.A.; Fraser, S.P.; Brackenbury, W.J. In Vivo Evidence for Voltage-Gated Sodium Channel Expression in Carcinomas and Potentiation of Metastasis. Cancers 2019, 11, 1675. [Google Scholar] [CrossRef] [Green Version]
  7. Kabra, R.; Singh, S. Transporter proteins and its implication in human diseases. Adv. Protein Chem. Struct. Biol. 2020, 124, 1–21. [Google Scholar] [CrossRef]
  8. Amawi, H.; Sim, H.-M.; Tiwari, A.K.; Ambudkar, S.V.; Shukla, S. ABC Transporter-Mediated Multidrug-Resistant Cancer. In Drug Transporters in Drug Disposition, Effects and Toxicity; Liu, X., Pan, G., Eds.; Spinger: Singapore, 2019; Volume 1141, pp. 549–580. [Google Scholar] [CrossRef]
  9. Nikolaou, M.; Pavlopoulou, A.; Georgakilas, A.G.; Kyrodimos, E. The challenge of drug resistance in cancer treat-ment: A current overview. Clin. Exp. Metastasis 2018, 35, 309–318. [Google Scholar] [CrossRef]
  10. Prevarskaya, N.; Skryma, R.; Shuba, Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol. Rev. 2018, 98, 559–621. [Google Scholar] [CrossRef] [Green Version]
  11. Abuammar, H.; Bhattacharjee, A.; Simon-Vecsei, Z.; Blastyák, A.; Csordás, G.; Páli, T.; Juhász, G. Ion Channels and Pumps in Autophagy: A Reciprocal Relationship. Cells 2021, 10, 3537. [Google Scholar] [CrossRef]
  12. Steinbrueck, A.; Sedgwick, A.C.; Brewster, J.T.; Yan, K.-C.; Shang, Y.; Knoll, D.M.; Vargas-Zúñiga, G.I.; He, X.-P.; Tian, H.; Sessler, J.L. Transition metal chelators, pro-chelators, and ionophores as small molecule cancer chemotherapeutic agents. Chem. Soc. Rev. 2020, 49, 3726–3747. [Google Scholar] [CrossRef]
  13. Rieber, M. Cancer Pro-oxidant Therapy Through Copper Redox Cycling: Repurposing Disulfiram and Tetrathiomolybdate. Curr Pharm Des. 2020, 26, 4461–4466. [Google Scholar] [CrossRef] [PubMed]
  14. Shi, C.; Yang, E.J.; Tao, S.; Ren, G.; Mou, P.K.; Shim, J.S. Natural products targeting cancer cell dependency. J. Antibiot. 2021, 74, 677–686. [Google Scholar] [CrossRef] [PubMed]
  15. Li, Y. Copper homeostasis: Emerging target for cancer treatment. IUBMB Life 2020, 72, 1900–1908. [Google Scholar] [CrossRef] [PubMed]
  16. Babak, M.; Ahn, D. Modulation of Intracellular Copper Levels as the Mechanism of Action of Anticancer Copper Complexes: Clinical Relevance. Biomedicines 2021, 9, 852. [Google Scholar] [CrossRef]
  17. Gaur, K.; Vázquez-Salgado, A.M.; Duran-Camacho, G.; Dominguez-Martinez, I.; Benjamín-Rivera, J.A.; Fernández-Vega, L.; Sarabia, L.C.; García, A.C.; Pérez-Deliz, F.; Román, J.A.M.; et al. Iron and Copper Intracellular Chelation as an Anticancer Drug Strategy. Inorganics 2018, 6, 126. [Google Scholar] [CrossRef] [Green Version]
  18. Oliveri, V. Biomedical applications of copper ionophores. Coord. Chem. Rev. 2020, 422, 213474. [Google Scholar] [CrossRef]
  19. Park, S.S.; Lee, D.M.; Lim, J.H.; Lee, D.; Park, S.J.; Kim, H.M.; Sohn, S.; Yoon, G.; Eom, Y.W.; Jeong, S.-Y.; et al. Pyrrolidine dithiocarbamate reverses Bcl-xL-mediated apoptotic resistance to doxorubicin by inducing paraptosis. Carcinogenesis 2018, 39, 458–470. [Google Scholar] [CrossRef]
  20. Cheng, X.; Li, D.; Sun, M.; He, L.; Zheng, Y.; Wang, X.; Tang, R. Co-delivery of DOX and PDTC by pH-sensitive nanoparticles to overcome multidrug resistance in breast cancer. Colloids Surf. B Biointerfaces 2019, 181, 185–197. [Google Scholar] [CrossRef]
  21. Li, H.; Wang, J.; Wu, C.; Wang, L.; Chen, Z.-S.; Cui, W. The combination of disulfiram and copper for cancer treatment. Drug Discov. Today 2020, 25, 1099–1108. [Google Scholar] [CrossRef]
  22. Pan, Q.; Xie, L.; Liu, R.; Pu, Y.; Wu, D.; Gao, W.; Luo, K.; He, B. Two birds with one stone: Copper metal-organic framework as a carrier of disulfiram prodrug for cancer therapy. Int. J. Pharm. 2022, 612, 121351. [Google Scholar] [CrossRef]
  23. Yang, Z.; Guo, F.; Albers, A.E.; Sehouli, J.; Kaufmann, A.M. Disulfiram modulates ROS accumulation and overcomes synergistically cisplatin resistance in breast cancer cell lines. Biomed. Pharmacother. 2019, 113, 108727. [Google Scholar] [CrossRef] [PubMed]
  24. Khan, H.J.; Rohondia, S.O.; Ahmed, Z.S.O.; Zalavadiya, N.; Dou, Q.P. Increasing opportunities of drug repur-posing for treating breast cancer by the integration of molecular, histological, and systemic approaches. In Drug Repurposing in Cancer Therapy; Academic Press: Cambridge, MA, USA, 2020; pp. 121–172. [Google Scholar]
  25. Kim, J.H.; Gupta, S.C.; Park, B.; Yadav, V.R.; Aggarwal, B.B. Turmeric (Curcuma longa) inhibits inflammatory nuclear factor (NF)-κB and NF-κB-regulated gene products and induces death receptors leading to suppressed proliferation, induced chemosensitization, and suppressed osteoclastogenesis. Mol. Nutr. Food Res. 2012, 56, 454–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Allensworth, J.L.; Evans, M.; Bertucci, F.; Aldrich, A.J.; Festa, R.A.; Finetti, P.; Ueno, N.T.; Safi, R.; McDonnell, D.P.; Thiele, D.J.; et al. Disulfiram (DSF) acts as a copper ionophore to induce copper-dependent oxidative stress and mediate anti-tumor efficacy in inflammatory breast cancer. Mol. Oncol. 2015, 9, 1155–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Peng, X.; Pan, Q.; Li, J.; Zhu, W.; Zhang, N.; Pu, Y.; Luo, K.; He, B. Polymer-directed supramolecular assembly of photo-sensitizers: Evocation of photothermal effect and highly efficient loading of disulfiram for chemo-phototherapy of cancer. Appl. Mater. Today 2021, 22, 100931. [Google Scholar] [CrossRef]
  28. Zhang, C.; Su, Z.-Y.; Khor, T.O.; Shu, L.; Kong, A.-N.T. Sulforaphane enhances Nrf2 expression in prostate cancer TRAMP C1 cells through epigenetic regulation. Biochem. Pharmacol. 2013, 85, 1398–1404. [Google Scholar] [CrossRef] [Green Version]
  29. Li, Y.; Chen, F.; Chen, J.; Chan, S.; He, Y.; Liu, W.; Zhang, G. Disulfiram/Copper Induces Antitumor Activity against Both Nasopharyngeal Cancer Cells and Cancer-Associated Fibroblasts through ROS/MAPK and Ferroptosis Pathways. Cancers 2020, 12, 138. [Google Scholar] [CrossRef] [Green Version]
  30. Tardito, S.; Bassanetti, I.; Bignardi, C.; Elviri, L.; Tegoni, M.; Mucchino, C.; Bussolati, O.; Franchi-Gazzola, R.; Marchiò, L. Copper Binding Agents Acting as Copper Ionophores Lead to Caspase Inhibition and Paraptotic Cell Death in Human Cancer Cells. J. Am. Chem. Soc. 2011, 133, 6235–6242. [Google Scholar] [CrossRef]
  31. Oliveri, V. Selective Targeting of Cancer Cells by Copper Ionophores: An Overview. Front. Mol. Biosci. 2022, 9, 841814. [Google Scholar] [CrossRef]
  32. Helsel, M.E.; Franz, K.J. Pharmacological activity of metal binding agents that alter copper bioavailability. Dalton Trans. 2015, 44, 8760–8770. [Google Scholar] [CrossRef] [Green Version]
  33. Pyun, J.; McInnes, L.E.; Donnelly, P.S.; Mawal, C.; Bush, A.I.; Short, J.L.; Nicolazzo, J.A. Copper bis (thiosemi-carbazone) complexes modulate P-glycoprotein expression and function in human brain microvascular endothelial cells. J. Neurochem. 2022. [Google Scholar] [CrossRef]
  34. Wang, X.; Yan, M.; Wang, Q.; Wang, H.; Wang, Z.; Zhao, J.; Li, J.; Zhang, Z. In Vitro DNA-binding, anti-oxidant and anticancer activity of indole-2-carboxylic acid dinuclear copper (II) complexes. Molecules 2017, 22, 171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wang, K.; Michelakos, T.; Wang, B.; Shang, Z.; DeLeo, A.B.; Duan, Z.; Hornicek, F.J.; Schwab, J.H.; Wang, X. Targeting cancer stem cells by disulfiram and copper sensitizes radioresistant chondrosarcoma to radiation. Cancer Lett. 2021, 505, 37–48. [Google Scholar] [CrossRef] [PubMed]
  36. Xu, Y.; Lu, L.; Luo, J.; Wang, L.; Zhang, Q.; Cao, J.; Jiao, Y. Disulfiram Alone Functions as a Radiosensitizer for Pancreatic Cancer Both In Vitro and In Vivo. Front. Oncol. 2021, 11, 683695. [Google Scholar] [CrossRef] [PubMed]
  37. Sabithakala, T.; Chittireddy, V.R.R. DNA binding and In Vitro anticancer activity of 2-((1H -benzimidazol-2-yl)methylamino)acetic acid and its copper(II) mixed-polypyridyl complexes: Synthesis and crystal structure. Appl. Organomet. Chem. 2018, 32, e4550. [Google Scholar] [CrossRef]
  38. Damelin, L.H.; Jivan, R.; Veale, R.B.; Rousseau, A.L.; Mavri-Damelin, D. Metformin induces an intracellular re-ductive state that protects oesophageal squamous cell carcinoma cells against cisplatin but not copper-bis (thiosemicarbazones). BMC Cancer 2014, 14, 314. [Google Scholar] [CrossRef] [Green Version]
  39. Seebacher, N.A.; Richardson, D.R.; Jansson, P.J. A mechanism for overcoming P-glycoprotein-mediated drug re-sistance: Novel combination therapy that releases stored doxorubicin from lysosomes via lysosomal permeabilization using Dp44mT or DpC. Cell Death Dis. 2016, 7, e2510. [Google Scholar] [CrossRef] [Green Version]
  40. Zhu, H.; Fan, J.; Wang, J.; Mu, H.; Peng, X. An “Enhanced PET”-Based Fluorescent Probe with Ultrasensitivity for Imaging Basal and Elesclomol-Induced HClO in Cancer Cells. J. Am. Chem. Soc. 2014, 136, 12820–12823. [Google Scholar] [CrossRef]
  41. Gao, W.; Huang, Z.; Duan, J.; Nice, E.C.; Lin, J.; Huang, C. Elesclomol induces copper-dependent ferroptosis in colorectal cancer cells via degradation of ATP7A. Mol. Oncol. 2021, 15, 3527–3544. [Google Scholar] [CrossRef]
  42. Ejaz, H.W.; Wang, W.; Lang, M. Copper toxicity links to pathogenesis of Alzheimer’s disease and therapeutics ap-proaches. Int. J. Mol. Sci. 2020, 21, 7660. [Google Scholar] [CrossRef]
  43. Bellia, F.; Grasso, G.I.; Ahmed, I.M.M.; Oliveri, V.; Vecchio, G. Carnoquinolines Target Copper Dyshomeostasis, Aberrant Protein–Protein Interactions, and Oxidative Stress. Chem. A Eur. J. 2020, 26, 16690–16705. [Google Scholar] [CrossRef]
  44. Bansal, R.; Suryan, A. A Comprehensive Review on Steroidal Bioconjugates as Promising Leads in Drug Discovery. ACS Bio. Med. Chem. Au. 2022. [Google Scholar] [CrossRef]
  45. Chen, X.; Nardon, C.; Dou, Q.P. Editorial: Unravelling Copper-Regulatory Systems and Copper-Affected Pathways in Cancer Cells to Improve Current Therapies. Front. Mol. Biosci. 2022, 9, 876902. [Google Scholar] [CrossRef] [PubMed]
  46. Bao, X.-Z.; Wang, Q.; Ren, X.-R.; Dai, F.; Zhou, B. A hydrogen peroxide-activated Cu(II) pro-ionophore strategy for modifying naphthazarin as a promising anticancer agent with high selectivity for generating ROS in HepG2 cells over in L02 cells. Free Radic. Biol. Med. 2020, 152, 597–608. [Google Scholar] [CrossRef] [PubMed]
  47. Oliveri, V.; Viale, M.; Caron, G.; Aiello, C.; Gangemi, R.; Vecchio, G. Glycosylated copper(ii) ionophores as prodrugs for β-glucosidase activation in targeted cancer therapy. Dalton Trans. 2013, 42, 2023–2034. [Google Scholar] [CrossRef]
  48. Prevarskaya, N.; Ouadid-Ahidouch, H.; Skryma, R.; Shuba, Y. Remodelling of Ca2+ transport in cancer: How it contributes to cancer hallmarks? Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130097. [Google Scholar] [CrossRef] [Green Version]
  49. Humeau, J.; Bravo-San Pedro, J.M.; Vitale, I.; Nuñez, L.; Villalobos, C.; Kroemer, G.; Senovilla, L. Calcium signaling and cell cycle: Progression or death. Cell Calcium 2018, 70, 3–15. [Google Scholar] [CrossRef]
  50. Wei, C.; Wang, X.; Chen, M.; Ouyang, K.; Song, L.-S.; Cheng, H. Calcium flickers steer cell migration. Nature 2009, 457, 901–905. [Google Scholar] [CrossRef] [Green Version]
  51. Visser, D.; Langeslag, M.; Kedziora, K.M.; Klarenbeek, J.; Kamermans, A.; Horgen, F.D.; Fleig, A.; van Leeuwen, F.N.; Jalink, K. TRPM7 triggers Ca2+ sparks and invadosome formation in neuroblastoma cells. Cell Calcium 2013, 54, 404–415. [Google Scholar] [CrossRef] [Green Version]
  52. Monteith, G.R.; Prevarskaya, N.; Roberts-Thomson, S.J. The calcium–cancer signalling nexus. Nat. Rev. Cancer 2017, 17, 373–380. [Google Scholar] [CrossRef] [Green Version]
  53. Chen, Y.-F.; Lin, P.-C.; Yeh, Y.-M.; Chen, L.-H.; Shen, M.-R. Store-Operated Ca2+ Entry in Tumor Progression: From Molecular Mechanisms to Clinical Implications. Cancers 2019, 11, 899. [Google Scholar] [CrossRef] [Green Version]
  54. Gagliardi, L.J.; Shain, D.H. Biophysical mechanism for zinc as an anticancer agent. Med. Hypotheses 2020, 144, 110273. [Google Scholar] [CrossRef] [PubMed]
  55. Xue, J.; Moyer, A.; Peng, B.; Wu, J.; Hannafon, B.N.; Ding, W.-Q. Chloroquine Is a Zinc Ionophore. PLoS ONE 2014, 9, e109180. [Google Scholar] [CrossRef] [Green Version]
  56. Huczyński, A. Polyether ionophores—promising bioactive molecules for cancer therapy. Bioorg. Med. Chem. Lett. 2012, 22, 7002–7010. [Google Scholar] [CrossRef] [Green Version]
  57. Rutkowski, J.; Brzezinski, B. Structures and Properties of Naturally Occurring Polyether Antibiotics. BioMed Res. Int. 2013, 2013, 162513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Dembitsky, V.M. Natural Polyether Ionophores and Their Pharmacological Profile. Mar. Drugs 2022, 20, 292. [Google Scholar] [CrossRef]
  59. MichałAntoszczak, J.R.; Huczynski, A. Structure and biological activity of polyether ionophores and their semisynthetic derivatives. In Bioactive Natural Products: Chemistry and Biology; Wiley-VCH Verlag GmbH & Co. KGaA, Boschstr. 12: Weinheim, Germany, 2015. [Google Scholar]
  60. Sulik, M.; Maj, E.; Wietrzyk, J.; Huczyński, A.; Antoszczak, M. Synthesis and Anticancer Activity of Dimeric Polyether Ionophores. Biomolecules 2020, 10, 1039. [Google Scholar] [CrossRef]
  61. Liu, F.; Li, W.; Hua, S.; Han, Y.; Xu, Z.; Wan, D.; Wang, Y.; Chen, W.; Kuang, Y.; Shi, J.; et al. Nigericin Exerts Anticancer Effects on Human Colorectal Cancer Cells by Inhibiting Wnt/β-catenin Signaling Pathway. Mol. Cancer Ther. 2018, 17, 952–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Xu, Z.; Shen, J.; Hua, S.; Wan, D.; Chen, Q.; Han, Y.; Ren, R.; Liu, F.; Du, Z.; Guo, X.; et al. High-throughput sequencing of circRNAs reveals novel insights into mechanisms of nigericin in pancreatic cancer. BMC Genom. 2019, 20, 716. [Google Scholar] [CrossRef] [Green Version]
  63. Bharti, H.; Singal, A.; Raza, M.; Ghosh, P.; Nag, A. Ionophores as Potent Anti-malarials: A Miracle in the Making. Curr. Top. Med. Chem. 2018, 18, 2029–2041. [Google Scholar] [CrossRef]
  64. Li, G.; De Oliveira, D.M.; Walker, M.J. The antimicrobial and immunomodulatory effects of Ionophores for the treatment of human infection. J. Inorg. Biochem. 2022, 227, 111661. [Google Scholar] [CrossRef]
  65. Hsu, C.G.; Chávez, C.L.; Zhang, C.; Sowden, M.; Yan, C.; Berk, B.C. The lipid peroxidation product 4-hydroxynonenal inhibits NLRP3 inflammasome activation and macrophage pyroptosis. Cell Death Differ. 2022, 1–14. [Google Scholar] [CrossRef] [PubMed]
  66. Zanke, B.W.; Lee, C.; Arab, S.; Tannock, I.F. Death of tumor cells after intracellular acidification is dependent on stress-activated protein kinases (SAPK/JNK) pathway activation and cannot be inhibited by Bcl-2 expression or interleukin 1β-converting enzyme inhibition. Cancer Res. 1998, 58, 2801–2808. [Google Scholar] [PubMed]
  67. Gao, G.; Liu, F.; Xu, Z.; Wan, D.; Han, Y.; Kuang, Y.; Wang, Q.; Zhi, Q. Evidence of nigericin as a potential therapeutic candidate for cancers: A review. Biomed. Pharmacother. 2021, 137, 111262. [Google Scholar] [CrossRef]
  68. Deng, C.-C.; Liang, Y.; Wu, M.-S.; Feng, F.-T.; Hu, W.-R.; Chen, L.-Z.; Feng, Q.-S.; Bei, J.-X.; Zeng, Y.-X. Nigericin selectively targets cancer stem cells in nasopharyngeal carcinoma. Int. J. Biochem. Cell Biol. 2013, 45, 1997–2006. [Google Scholar] [CrossRef] [PubMed]
  69. Gupta, P.B.; Onder, T.T.; Jiang, G.; Tao, K.; Kuperwasser, C.; Weinberg, R.A.; Lander, E.S. Identification of Selective Inhibitors of Cancer Stem Cells by High-Throughput Screening. Cell 2009, 138, 645–659. [Google Scholar] [CrossRef] [Green Version]
  70. Naujokat, C.; Fuchs, D.; Opelz, G. Salinomycin in cancer: A new mission for an old agent. Mol. Med. Rep. 2010, 3, 555–559. [Google Scholar] [CrossRef]
  71. Lu, Y.; Zhang, C.; Li, Q.; Mao, J.; Ma, W.; Yu, X.; Hou, Z.; Li, L. [Inhibitory effect of salinomycin on human breast cancer cells MDA-MB-231 proliferation through Hedgehog signaling pathway]. Zhonghua Bing Li Xue Za Zhi = Chin. J. Pathol. 2015, 44, 395–398. [Google Scholar]
  72. Fu, Y.-Z.; Yan, Y.-Y.; He, M.; Xiao, Q.-H.; Yao, W.-F.; Zhao, L.; Wu, H.-Z.; Yu, Z.-J.; Zhou, M.-Y.; Lv, M.-T.; et al. Salinomycin induces selective cytotoxicity to MCF-7 mammosphere cells through targeting the Hedgehog signaling pathway. Oncol. Rep. 2016, 35, 912–922. [Google Scholar] [CrossRef] [Green Version]
  73. Lu, Y.; Ma, W.; Mao, J.; Yu, X.; Hou, Z.; Fan, S.; Song, B.; Wang, H.; Li, J.; Kang, L.; et al. Salinomycin exerts anticancer effects on human breast carcinoma MCF-7 cancer stem cells via modulation of Hedgehog signaling. Chem. Interact. 2015, 228, 100–107. [Google Scholar] [CrossRef]
  74. Angeloni, V.; Tiberio, P.; Appierto, V.; Daidone, M.G. Implications of stemness-related signaling pathways in breast cancer response to therapy. Semin. Cancer Biol. 2015, 31, 43–51. [Google Scholar] [CrossRef]
  75. Dewangan, J.; Srivastava, S.; Mishra, S.; Divakar, A.; Kumar, S.; Rath, S.K. Salinomycin inhibits breast cancer progression via targeting HIF-1α/VEGF mediated tumor angiogenesis In Vitro and In Vivo. Biochem. Pharmacol. 2019, 164, 326–335. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, Y.; Liu, L.; Li, F.; Wu, T.; Jiang, H.; Jiang, X.; Du, X.; Wang, Y. Salinomycin Exerts Anticancer Effects on PC-3 Cells and PC-3-Derived Cancer Stem Cells In Vitro and In Vivo. BioMed Res. Int. 2017, 2017, 4101653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Dewangan, J.; Srivastava, S.; Rath, S.K. Salinomycin: A new paradigm in cancer therapy. Tumor Biol. 2017, 39, 1010428317695035. [Google Scholar] [CrossRef] [Green Version]
  78. Jiang, J.; Li, H.; Qaed, E.; Zhang, J.; Song, Y.; Wu, R.; Bu, X.; Wang, Q.; Tang, Z. Salinomycin, as an autophagy modulator-- a new avenue to anticancer: A review. J. Exp. Clin. Cancer Res. 2018, 37, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Xu, G.; Tang, K.; Hao, Y.; Wang, X.; Sui, L. Polymeric Nanocarriers Loaded with a Combination of Gemcitabine and Salinomycin: Potential Therapeutics for Liver Cancer Treatment. J. Clust. Sci. 2022, 1–12. [Google Scholar] [CrossRef]
  80. Li, R.; Guo, N.; Fu, L.; & Miao, Y. A Feasible Strategy of Fabricating Redox-Responsive Polymeric Salinomycin Small Molecule Prodrug Delivery for Liver Cancer Therapy. J. Clust. Sci. 2022, 1–11. [Google Scholar] [CrossRef]
  81. Venkatadri, R.; Iyer, A.K.V.; Kaushik, V.; Azad, N. A novel resveratrol–salinomycin combination sensitizes ER-positive breast cancer cells to apoptosis. Pharmacol. Rep. 2017, 69, 788–797. [Google Scholar] [CrossRef]
  82. Antoszczak, M. A comprehensive review of salinomycin derivatives as potent anticancer and anti-CSCs agents. Eur. J. Med. Chem. 2019, 166, 48–64. [Google Scholar] [CrossRef]
  83. Naujokat, C.; Steinhart, R. Salinomycin as a Drug for Targeting Human Cancer Stem Cells. J. Biomed. Biotechnol. 2012, 2012, 950658. [Google Scholar] [CrossRef]
  84. Muntimadugu, E.; Kumar, R.; Saladi, S.; Rafeeqi, T.A.; Khan, W. CD44 targeted chemotherapy for co-eradication of breast cancer stem cells and cancer cells using polymeric nanoparticles of salinomycin and paclitaxel. Colloids Surf. B Biointerfaces 2016, 143, 532–546. [Google Scholar] [CrossRef]
  85. Qi, D.; Liu, Y.; Li, J.; Huang, J.H.; Hu, X.; Wu, E. Salinomycin as a potent anticancer stem cell agent: State of the art and future directions. Med. Res. Rev. 2022, 42, 1037–1063. [Google Scholar] [CrossRef]
  86. Erdogan, S.; Serttas, R.; Turkekul, K.; Dibirdik, I. The synergistic anticancer effect of salinomycin combined with cabazitaxel in CD44+ prostate cancer cells by downregulating wnt, NF-κB and AKT signaling. Mol. Biol. Rep. 2022, 49, 4873–4884. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, H.; Zhang, H.; Zhu, Y.; Wu, Z.; Cui, C.; Cai, F. Anticancer Mechanisms of Salinomycin in Breast Cancer and Its Clinical Applications. Front. Oncol. 2021, 11, 654428. [Google Scholar] [CrossRef] [PubMed]
  88. Zhao, Y.-X.; Ho, C.-K.; Xie, Y.; Chen, Y.-H.; Li, H.-Z.; Zhang, G.-Y.; Li, Q.-F. Calcimycin Suppresses S100A4 Expression and Inhibits the Stimulatory Effect of Transforming Growth Factor β1 on Keloid Fibroblasts. Ann. Plast. Surg. 2018, 81, 163–169. [Google Scholar] [CrossRef] [PubMed]
  89. Shah, A.M.; Wani, A.; Qazi, P.H.; Rehman, S.-U.; Mushtaq, S.; Ali, S.A.; Hussain, A.; Shah, A.; Qazi, A.K.; Makhdoomi, U.S.; et al. Isolation and characterization of alborixin from Streptomyces scabrisporus: A potent cytotoxic agent against human colon (HCT-116) cancer cells. Chem. Interact. 2016, 256, 198–208. [Google Scholar] [CrossRef] [PubMed]
  90. Markowska, A.; Kaysiewicz, J.; Markowska, J.; Huczyński, A. Doxycycline, salinomycin, monensin and ivermectin repositioned as cancer drugs. Bioorg. Med. Chem. Lett. 2019, 29, 1549–1554. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of metallic and polyether ionophores.
Figure 1. Chemical structure of metallic and polyether ionophores.
Molecules 27 04708 g001
Figure 2. Anti-cancerous potential of nigericin with their associated mechanism.
Figure 2. Anti-cancerous potential of nigericin with their associated mechanism.
Molecules 27 04708 g002
Figure 3. Anti-cancerous potential of salinomycin with their associated mechanism.
Figure 3. Anti-cancerous potential of salinomycin with their associated mechanism.
Molecules 27 04708 g003
Table 1. Numerous classes of Copper (Cu) ionophores in cancer therapeutics.
Table 1. Numerous classes of Copper (Cu) ionophores in cancer therapeutics.
Classes of Cu IonophoresAnticancer Mode of ActionChemical StructureReference
Cu-Pyrrolidine dithiocarbamate (PDTC)
  • Transport redox-active Cu into the cell
  • Inhibits the cell proliferation, nuclear factor-jB (NF-jB), and ubiquitin–proteasome system inducing apoptosis
Molecules 27 04708 i001[34]
Cu-DSF (Disulfiram)
  • Targets stem-like cancer cells and augment the cytotoxic efficacies of several anticancer drugs
  • Inhibited NF-jB up-regulation in ROS-targeted chemotherapeutic approaches.
  • Decreased GSH levels
Molecules 27 04708 i002[35,36]
Cu-DPy (disulfide-based Cu carrier, 2,20
-dithiodipyridine)
  • ROS induction
  • Cytotoxic effects against various cancer cells
  • Increases intracellular Cu concentration
  • Acts as a recyclable Cu ionophore
  • Affect thioredoxin and GSH system
Molecules 27 04708 i003[37]
Cu-GTSM and Cu-ATSM
  • Possess different reduction potential and cell metabolism
  • Inhibits the chymotrypsin-like activity of the proteasome
  • ROS generation
  • Lysosomal membrane permeabilization
  • cathepsin release
  • Apoptotic cascades
Molecules 27 04708 i004[38]
Cu- Dp44mT (Di-2-pyridylketone-4,4,-dimethyl-3-thiosemicarbazone
) + Dpc (di-2-pyridylketone 4-methyl-4-cyclohexyl-3-thiose
micarbazone)
  • Improved anticancer activity
  • Reduced toxicity
  • Increase in intracellular Cu concentration
  • ROS production
  • Apoptosis induction
Molecules 27 04708 i005[39]
Elesclomol
  • Completed a series of clinical trials for patients with advanced melanoma, acute myeloid leukemia, ovarian epithelial cancer, fallopian tube cancer, and primary peritoneal
  • cancer, prostate cancer, and other solid tumors
  • Oxidative stress induction
  • Apoptosis induction
  • Promotes the increase of intracellular Cu levels
  • ROS generation
  • Exert cytotoxicity through DNA-damage
  • Regulates ferroptosis
Molecules 27 04708 i006[40,41]
Cu-CQ (7-iodo-5-chloro-8-hydroxyquinoline)
  • Cell death induction
  • Increased cytotoxicity in combination
  • Increase in intracellular Cu
  • Inhibit the proteasome activity
  • Cytoplasmic XIAP clearance (anti-apoptotic protein) that inhibits caspases activation
  • Apoptosis induction
Molecules 27 04708 i007[31]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pandey, P.; Khan, F.; Qari, H.A.; Upadhyay, T.K.; Alkhateeb, A.F.; Oves, M. Evidence of Metallic and Polyether Ionophores as Potent Therapeutic Drug Candidate in Cancer Management. Molecules 2022, 27, 4708. https://doi.org/10.3390/molecules27154708

AMA Style

Pandey P, Khan F, Qari HA, Upadhyay TK, Alkhateeb AF, Oves M. Evidence of Metallic and Polyether Ionophores as Potent Therapeutic Drug Candidate in Cancer Management. Molecules. 2022; 27(15):4708. https://doi.org/10.3390/molecules27154708

Chicago/Turabian Style

Pandey, Pratibha, Fahad Khan, Huda A. Qari, Tarun Kumar Upadhyay, Abdulhameed F. Alkhateeb, and Mohammad Oves. 2022. "Evidence of Metallic and Polyether Ionophores as Potent Therapeutic Drug Candidate in Cancer Management" Molecules 27, no. 15: 4708. https://doi.org/10.3390/molecules27154708

Article Metrics

Back to TopTop