Next Article in Journal
Comprehensive Analysis of Germline Variants in Mexican Patients with Hereditary Breast and Ovarian Cancer Susceptibility
Next Article in Special Issue
Sensitization of Cancer Cells to Radiation and Topoisomerase I Inhibitor Camptothecin Using Inhibitors of PARP and Other Signaling Molecules
Previous Article in Journal
The Cooperative Relationship between STAT5 and Reactive Oxygen Species in Leukemia: Mechanism and Therapeutic Potential
Previous Article in Special Issue
NGAL is Downregulated in Oral Squamous Cell Carcinoma and Leads to Increased Survival, Proliferation, Migration and Chemoresistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ionophores: Potential Use as Anticancer Drugs and Chemosensitizers

1
Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA
2
Great Plains Health, North Platte, NE 69101, USA
*
Author to whom correspondence should be addressed.
Cancers 2018, 10(10), 360; https://doi.org/10.3390/cancers10100360
Submission received: 23 August 2018 / Revised: 13 September 2018 / Accepted: 21 September 2018 / Published: 27 September 2018
(This article belongs to the Special Issue Sensitization Strategies in Cancer Treatment)

Abstract

:
Ion homeostasis is extremely important for the survival of both normal as well as neoplastic cells. The altered ion homeostasis found in cancer cells prompted the investigation of several ionophores as potential anticancer agents. Few ionophores, such as Salinomycin, Nigericin and Obatoclax, have demonstrated potent anticancer activities against cancer stem-like cells that are considered highly resistant to chemotherapy and responsible for tumor relapse. The preclinical success of these compounds in in vitro and in vivo models have not been translated into clinical trials. At present, phase I/II clinical trials demonstrated limited benefit of Obatoclax alone or in combination with other anticancer drugs. However, future development in targeted drug delivery may be useful to improve the efficacy of these compounds. Alternatively, these compounds may be used as leading molecules for the development of less toxic derivatives.

1. Introduction

Ion Transport

Ion homeostasis is extremely important for the survival of cells. A proper balance of ions both inside the cells as well as in the extracellular matrix is necessary for the maintenance of membrane potential, cell shape and proper functioning of several cellular pathways. Cell membrane consists of two lipid bi-layers where polar heads of the lipids face outward and hydrophobic tails form interior of the cell membrane. This structural composition of plasma membrane results in impermeability of various ions, small hydrophilic molecules such as glucose and macromolecules such as proteins and RNA across the cell membrane. Only water, oxygen and carbon dioxide freely move across plasma membrane. Cells overcome these transport issues by devising mechanisms for facilitated diffusion as well as active transport of ions and molecules across membrane. Facilitated transport involves diffusion of ions towards concentration gradient mediated by proteins which form water filled ion channels across the membrane. These ion channels are gated and can be opened and closed based on the cellular requirements. The most common types of gated ion channels are ligand-gated, mechanically gated, voltage-gated, and light-gated [1]. In active transport ions or molecules are transported against the concentration gradient with the help of transporter proteins using energy from the ATP. Na+/K+ ATPase, H+/K+ ATPase, Ca2+ ATPase, ABC transporters are a few examples of active transporters [2]. Aberrant expression and/or functioning of ion channels and ion pumps in cancer cells establish a unique ion homeostasis which is specifically advantageous to cancer cells. Maintenance of this ion homeostasis is of great interest to cancer cells. In following sections of the review article, we will discuss how cancer cells maintain a particular ion balance and how it helps them in escaping death, increased cancer cell proliferation, and metastasis. We will also explore how this ion homeostasis is targeted to develop novel therapeutic interventions for cancer treatment and discuss role of ionophores as anticancer drugs. Ionophores are a class of compounds which have been successfully employed to eliminate cancer by manipulating ion balance in cancer. We will focus on Salinomycin (SAL), Nigericin (NIG) and Obatoclax (OBT) as these ionophores have shown well-documented potent anticancer activity against cancer stem-like cells as well as promising use as chemosensitizer. Although other ionophores have shown anticancer activity and some of them may be active against putative cancer stem cells (CSCs) the information is limited or they are less promising than SAL, NIG or OBT. For instance, Valinomycin’s activity against cancer cells were less potent compared to SAL or NIG [3]. (Table 1) We will also track clinical progress of these drugs by evaluating various clinical trials exploiting these drugs as potential anticancer agents.

2. Ion Transport in Cancer Cells and Its Targeting to Develop Novel Anticancer Therapies

2.1. Ion Transport in Cancer Cells

Growing body of evidence suggest an altered ion transport in cancer cells. Cancer cells rewire their cellular circuitry to establish, adopt, proliferate, and metastasize in various challenging conditions by manipulation their ion homeostasis and ion channels and ion pumps play a critical role in this reorganization [30].
Ca2+ is a very important ion and plays key role in various signaling mechanism, which integrate with other signal-transduction cascades and controls a variety of cellular processes. Therefore, intracellular Ca2+ (Ca2+i) concentration is precisely maintained for proper functioning of cells. Ca2+ homeostasis is maintained by calcium permeable channels such as transient receptor potential (TRP) channels, store-operated channels (SOCs), voltage-gated calcium channels, as well as mitochondrial calcium uniporter (MCU), voltage-dependent anion channels (VDACs), IP3 and ryanodine receptors, and others. Ca2+i plays central role in early G1phase and at the G1/S and G2/M transitions [31]. Other studies have pointed towards Ca2+/calmodulin (CaM) and Ca2+/calcineurin pathways as major checkpoints in cell cycle progression [32,33]. Metastasis involves several Ca2+ dependent processes, including cell deformation, invasion, migration, and adhesion. TRPM7 channels have been shown to form local and transient calcium domains known as “calcium flickers” at lamellipodia and guide the direction of migration [34]. An in-depth account of Ca2+ transport mediated proliferation and metastasis in prostate, breast and lung cancer can be found in a review article by Deliot et al. [35] Apoptosis involving calcium ion overload in cytosol is a well explored process. Apoptotic cells increase intrinsic Ca2+ either by sustained Ca2+ influx via activated channels or release of calcium by a stressed endoplasmic reticulum (ER) [36,37]. Cancer cells exhibit greater apoptotic resistance by inhibiting calcium influx by down-regulating channels and/or adapt to chronic-reduced ER Ca2+ [36].
K+ is the most predominant ion inside the cell and its cellular concentration is maintained by four different classes of ion channels―voltage-gated, calcium-activated, inward rectifier and two-pore-domain potassium channels. K+ ion is key regulator of cell volume and shrinking of cell volume is a characteristic phenotypic modulation of cells undergoing apoptosis. Loss of intracellular K+ via activation of various ion channels leads to apoptosis by decay of the membrane potential and the associated Ca2+ influx, apoptotic volume decrease, and activation of various enzymes involved in the apoptotic process [38]. Readers can refer to a review by Wang for the potential role played by K+ ion channels in cancer cell proliferation and apoptosis [39]. Similarly, Na+ and Cl ion and ion channels have been associated with cancer proliferation and resistance to apoptosis [38,40]. Zn2+ transport has been shown to affect epithelial mesenchymal transition (EMT) and metastatic stature of cancer [41]. Zn2+ accumulation has been linked to increased resistance as well as sensitization of cancer in cell dependent manner [42,43]. Mg2+ homeostasis is often associated with drug resistance of cancer cells [44,45]. Cu+ is essential micro nutrient and its levels are maintained by several transporter and chaperone proteins. Cu+ is selectively taken up by Copper Transport Protein 1 (CTR1) and then distributed inside the cell to various cellular compartments by three chaperone proteins Atox1, Cox17p and CCS. While Cox17p and CCS transport Cu+ to mitochondria and copper/zinc superoxide dismutase respectively; Atox1 carries Cu+ to ATPase transporters ATP7A and ATP7B which move Cu+ to trans-golgi network or secretory vesicles for copper efflux from the cells [46]. Pt2+ exhibits similar coordination chemistry that of Cu+ and highjack copper transport machinery for transport of platinum-based anticancer drugs. Modulation of expression and activity of CTR1, ATP7A and ATP7B has been shown to impart resistance to platinum-based chemotherapies by reducing their cellular levels either by inhibiting their uptake or increased efflux of internalized drug respectively [47,48,49,50].
Cancer cells switch their metabolism to glycolysis to meet their energy requirements, known as Warburg effect. This metabolic change leads to accumulation of lactate in the cells. This excess lactate is pumped outside the cell by the over activation of Na+/H+ exchanger 1 (NHE1) and the H+/lactate cotransporter in cancer cells making tumor microenvironment highly acidic. This deregulated pH homeostasis results in cellular alkalization which was suggested to be first step in the carcinogenesis [51]. Cancer cells use this acidic environment to their advantage as they are more adept to these conditions than normal cells. Furthermore, this acidic environment shields cancer from weakly basic drugs by protonating them resulting in their decreased partitioning inside cancer cells [52]. Alternatively, H+ ion channels and pumps play a key role in malignant transformation of cancer by inducing metastasis. Cancer metastasis involves cell volume and cell shape modifications as cell develops lamellipodium and invadopodium to migrate and invade through extracellular matrix. These changes in cell volume are regulated by local ion transport through the ion channels at the leading edge and tip of the outgrowing lamellipodium/invadopodium. Several studies have pointed towards a critical role played by NHE1 in tumor progression and invasion [53]. H+ ion channels also play important role in cell signaling, proliferation and cell cycle.

2.2. Ion Transport and Chemotherapy

Modulation and maintenance of altered ionic homeostasis by cancer cells is well documented. Therefore, alteration of this ionic balance may serve as a lynch pin to start a cascade of signaling events ultimately leading to cancer cell death. There is multitude of evidence to support this notion as several studies indicated modulation of cellular ion homeostasis by either activation or deactivation of ion transporters and ion channels sensitizes cancer cells to otherwise in effective drugs. Regarding their potential use as chemosensitizers, Abdoul-Azize et al. recently demonstrated sensitization of pediatric acute myloid leukemia to dexamethasone by chelation of intracellular Ca2+ ions with calcium chelator BAPTA-AM [54]. Similarly, phenyl isothiocyante induced apoptosis in Gefitinib-resistant NCI-H460 human lung cancer cells by altering Ca2+i levels [55]. Chen et al. reported reversal of chemoresistance to platinum drugs in oxaliplatin-resistant human cervical cancer cells in vitro and in vivo in response to co-treatment with iron chelator desferal [56]. Partial restoration of function of volume-sensitive outwardly rectifying (VSOR) chloride channels in cisplatin resistant KCP-4 human epidermoid cancer cell line by treatment with trichostatin A sensitized these cells to cisplatin [57]. Esomeprazole a proton pump inhibitor potentiated antitumor activity of doxorubicin in triple negative MDA-MB-468 breast cancer cells [58]. Specific ion pump inhibitors have been tested for cancer treatment. For example, cardiac glycosides which are Na+/K+ pump inhibitors and commonly used for heart ailments are currently being investigated for their anticancer properties in various cancer. Several studies have pointed towards their potential anticancer efficacy in different types of cancers [59,60,61,62]. Similarly, hERG1 inhibitors, which block kv11.1 channels, have shown remarkable antitumor activity in several cancers [63,64]. Even though, ion channel/ion pump inhibitors have demonstrated potent anticancer activity, their use as anticancer drugs is limited due to limited selectivity. For instance, cardiac glycosides have very narrow therapeutic index (low nano molar range) as at higher concentrations they exhibit severe cardio toxic side effects. Nonspecific inhibition of kv11.1 channels by hERG1 inhibitors may also lead to the lengthening of the electrocardiographic QT interval, thus predisposing the patient to ventricular arrhythmias [65]. Ion transport inhibitors may have other effects that can be exploited as chemosensitizer. For instance, the Ca++ channel inhibitor Verapamil is also a potent inhibitor of the multidrug resistant (MDR) protein ABCB1/P-glycoprotein (P-gp) and this property has been tested to overcome multidrug resistance [66]. Since the P-glycoprotein, can be modulated by calcium channel blockers such as cyclosporin and nifedipine these inhibitors were able to reverse drug resistance in tumors [67,68]. However, at present the use of calcium channel blockers in chemotherapy has not been successfully translated into clinics, perhaps to limited availability and toxicity. For instance, while the average steady-state plasma levels measured for verapamil is ~0.5 μM the concentration usually associated with MDR1 inhibition is about ≥50 µM [66] indicating that this drug may not be useful at the clinical level.
Therefore new, more specific and selective ion transport modulators are needed to effectively exploit cancer’s ionic homeostasis as a target to develop successful therapy interventions against cancer.

3. Ionophores

Ionophores mean “ion carrier” is a class of compounds which can bind non-covalently with ions and can assist in their transport across the cell membrane. Ionophore consists of lipophilic exterior and has a hydrophilic interior where ion binds and transported across lipid membrane. Ionophores can be divided in two categories depending on the size of the molecule that in turn determines the mode of transport of the ion across the cell membrane (Figure 1). Small ionophores such as valinomycin form complex with the ions and transport them across the cell membrane are known as “Ion Carriers” [69]. Small polyether ionophores transport ions by electroneutral, electrogenic or biomimetic mechanisms depending on micro environmental and structural modification of ionophore [70,71,72]. Large ionophores such as gramicidin form channels across the cell membrane for the ion transport. The interior of channel is hydrophilic and assists in ion transport while lipophilic external shell shields ion from hydrophobic environment of cell membrane [69]. However, ion channel ionophores exhibit a lower selectivity in comparison with ion carrier ionophores. Based on chemical structure ionophores can be categorized as polyether, peptide, cyclodepsipeptide, macrotetrolides and cryptates [69]. Polyether ionophores consist of an oxygen rich hydrophilic interior which mostly comprises of ether bonds. Salinomycin and nigericin are example of polyether ionophores. Gramicidin is example of peptide ionophores where peptide bonds contribute towards ionophortic activity of the peptide. Cyclodepsipeptides are peptides in which one or more peptide bonds are replaced by ester bonds. Enniantin is an example of this class of ionophores. Nonactin is a macrotetrolide and has a macrocyclic structure comprising of four tetrahydrofuranyl-ester residues. Commonly, ionophores have been used as anticoccidial drugs for poultry and/or growth promoters in ruminants. Lately, ionophores have found a new role as anticancer drugs due to their ability to alter this ion balance by ion transport across cell membrane.

3.1. Salinomycin (SAL)

SAL is a naturally occurring polyether antibiotic [73] isolated from Streptomyces albus strain. It has been used as antibacterial and coccidiostat. SAL is an ionophore and imparts its antibacterial properties by facilitating transport of K+ ions through the cell membrane of target organism leading to an increase of intracellular Ca2+ ions. This disruption of ionic homeostasis leads to deregulation of osmotic balance resulting in death of the organism. SAL came in forefront as a potential anticancer drug in 2009 when Gupta et al. screened roughly 16,000 compounds for their selective anticancer efficacy against CSCs and found SAL at least 100-fold more effective than paclitaxel, a commonly used anticancer drug [7]. Following this work, several other studies pointed towards SAL’s selectivity in targeting cancer stem cells practically in every type of cancer [74,75,76,77,78,79,80] as well as other multidrug resistance (MDR) cancer cells [81,82]. Although the exact mechanism by which SAL targets cancers is not known, it is very clear that it influences multiple pathways to impart its effects. SAL has been shown to induce cancer and CSC death by inducing apoptosis [83,84,85,86,87,88]. There are studies which indicate autophagic cancer cell death by SAL [89,90]. However, several studies present a contradictory view as they suggest inhibition of autophagy and induction of apoptosis as the mechanism for elimination of cancer by SAL [91,92,93,94,95]. Several studies indicated SAL induced oxidative stress as a key mediator for apoptotic cell death [84,96,97,98]. SAL induces oxidative stress by altering mitochondrial membrane potential. Beside its specific cytotoxicity towards cancer and CSCs SAL regulates cancer metastasis by inhibiting cancer cell invasion and migration by targeting Wnt and EMT pathways [78,99,100,101,102]. Several studies indicate possible involvement of Hedgehog signaling in SAL induced cell death in breast cancer [103,104]. SAL is a potent partner in a co-therapy approach and has been shown to sensitize several cancers and to potentiate efficacy of other commonly used anticancer drugs such as doxorubicin, trastuzumab, gemcitabine, tamoxifen etc. [105,106,107,108] Zhang et al. demonstrated SAL induced sensitization of pancreatic cancer to gemcitabine by targeting CSCs [108]. In a recent study Venkatadri et al., observed a sensitizing effect of SAL in breast cancer where SAL was able to potentiate the resveratrol’s anticancer effect at low concentration which was rather ineffective when used independently [109].
Cancer resistance is a major concern in complete eradication of cancer as some of the cancer cells acquire or exhibit resistance to therapy and escape elimination. These cells come back as a more aggressive and more resistance cancer and cause a cancer relapse. Most of the current therapeutic approaches fall short of achieving complete cure of cancer and after initial remission cancer relapses in several cases. Interestingly, SAL has demonstrated potent anticancer effect in various multi drug resistant (MDR) cancers [110,111,112]. Development of drug efflux mechanisms via various drug transporter proteins such as p-glycoproteins, ABCG, MDR etc. is the most commonly employed counter measure by cancer to survive a fatal outcome. SAL has been shown to overcome drug resistance by inhibiting these drug transporters [105,113,114].
SAL has exhibited a great therapeutic potential as an anticancer drug. However, poor water solubility and toxicity to normal cells is a major concern in its therapeutic application for cancer treatment. These issues can be addressed by either development of targeted delivery strategies and/or by synthesis of less toxic and more specific SAL analogues. Recently, several studies have reported successful use of nanoparticles, nanomicelles, nanotubes, and multilamellar liposomes conjugated with cancer cell surface markers such as CD133, CD44 etc. for targeted delivery of salinomycin [115,116,117,118,119]. Many analogues have been synthesized by introducing different substituent, functional groups in the core structure of salinomycin and have been tested for their anticancer efficacy [120,121]. SAL demonstrated unique ability to target CSCs and MDR cancer cells in variety of cancers bestowing it with enormous potential to be a break through drug as a mono therapy or as a sensitizer to compound the effects of other anticancer agents in resistant cancers.

3.2. Nigericin (NIG)

NIG is an antibiotic derived from Streptomyces Hygroscopicus. It was first isolated by Harned et al. from an unidentified Streptomyces which was later reported to be Streptomyces “Nig-1” [122,123]. NIG is an ionophore and can transport K+, H+ and Pb2+ ions across the plasma membrane. NIG is an antiporter of H+ and reduces internal pH (pHi) of cells. Cell proliferation is a pH sensitive process and DNA replication requires slightly alkaline pH to perform optimally. Several studies have indicated stimulation of cell growth resulting from rapid increase of pHi (0.1–0.3 pH units) in response to addition of growth promoting reagents such as serum and growth factors [124]. This increase in pHi in response to growth factors was mediated by stimulation of amiloride sensitive Na+/H+ exchange [125]. Cancer cells have a reversed pH gradient then normal cells which have pHe > pHi while cancer cell maintain a pHe < pHi. This reversed pH gradient is advantageous to cancer cells as it promotes cancer progression by inducing cancer invasion and migration [126]. NIG has shown potent anticancer potential in several cancers as well as resistant cancer stem cells [5,6]. Exact mechanism by which nigericin acts is not known but several studies have pointed towards its ability to antiport H+ inside the cell as possible mode for its anticancer activity. Cancer cells have an acidic external pH (pHe) and nigericin exploits these external cancer microenvironment conditions to its advantage by transporting H+ from exterior to interior of the cell resulting in lowering of pHi which in turn leads to cancer cell death. Margolis et al. reported a decrease in cytoplasm pH and reduction in DNA synthesis on treatment with millimolar concentrations of NIG in Ehrlich ascites tumor cells [127]. Alteration of mitochondrial function has been associated with anticancer activity of NIG. Treatment with NIG alters membrane potential which may lead to disruption of energy balance and/or increased reactive oxygen species (ROS) production leading to cancer cell death [3,128].
Photodynamic therapy (PDT) is a form of phototherapy which is minimally invasive and least toxic. It employs a light source which excites the photo sensitizer which in turn interacts with molecular oxygen to produces radicals and ROS leading to localized cytotoxicity in the exposed tissue. Several studies have pointed towards NIG mediated sensitization of cancer cells for photodynamic therapy [129,130]. There is no clear mechanistic explanation for this synergistic effect as evidence suggest change of pHi, inhibition of peroxidase detoxification to translocation of Bax to mitochondria as possible reasons for this outcome [131,132]. In another study, Varnes et al. observed an inhibition of potentially lethal damage recovery in A549 cells upon treatment with micro molar concentration of NIG post PDT [133]. NIG sensitizes cancer cells and demonstrated a synergistic effect with other anticancer drugs leading to an improved efficacy of these drugs [6,134]. This enhanced efficacy was mostly related to NIG’s ability to reduce pHi under acidic tumor microenvironment [134,135]. Regulation of key cellular pathways such as EMT and Wnt/β catenin has been shown to induce cytotoxicity and inhibition of cancer metastasis on NIG treatment [136,137]. Autophagy inhibition potentiates NIG treatment. In a recent study Vu et al. demonstrated a reduction in spheroid formation by ATG5 deficient glioma cells on treatment with NIG [138].

3.3. Obatoclax (OBT)

OBT is synthetic derivative of Prodigiosin class of compounds. Prodigiosin are red pigments produced by bacteria and show antimalarial, antifungal, immunosuppressant, antibiotic, and anticancer properties. OBT is a BH3-mimitic and induces apoptosis by modulating Bcl2 family proteins. It has shown cytotoxicity in several cancers by inducing apoptotic cell death.
Often faced with harsh conditions such as nutrient deficiency, hypoxia, DNA damage, unnatural genetic variation, and instability, cancer cells still manage to survive by evading death signals by modulating pro-survival regulators. One such survival design includes over expression of anti-apoptotic Bcl2 family proteins. These proteins antagonize pro-death proteins by forming a heterodimer via binding to the pro-apoptotic protein’s BH3 domain situated in the hydrophobic cleft of anti-apoptotic proteins. While pro-death proteins have only BH3 homology domain, Bcl2 proteins have four homology domains (BH1-4). Other apoptotic proteins Bax and Bak which are necessary for the cell death by BH3 only proteins share three homology domains (BH1-3). In viable cells, Bax and Bak exist as monomers; however, they form homooligomers upon activation of BH3-only pro-apoptotic proteins. These homooligomers insert themselves in mitochondrial membrane and permeabilize it for the release of cytochrome c and other pro-apoptotic factors needed for mitochondria mediated apoptotic cell death. Therefore, natural or synthetic BH3 mimics represent a promising therapeutic approach for targeting cancer cells.
OBT’s anticancer potential is often attributed to its BH3 targeted apoptosis induction in cancer cells. In a recent study, Díaz de Greñu et al. demonstrated ionophoretic activity of OBT and other synthetic derivatives towards chloride and bicarbonate anions and correlated cytotoxicity of these compounds for small-cell lung carcinoma cell line GLC4 with their ability to discharge pH gradient in living cells [139]. Acridine orange (AO) a membrane permeable dye which accumulates in acidic vesicles such as lysosomes was used to monitor pH changes with OBT treatment. AO have a characteristic orange fluorescence at acidic pH while it flourishes green at basic pH. A transition from orange to green fluorescence for AO was observed upon treatment with OBT indicating a possible bicarbonate ion transport leading to alkalization of lysosomes by OBT.
Even though apoptotic cell death constitutes the primary mode of cytotoxicity of OBT in cancer cells, other cell death mechanisms such as autophagy, necrosis, necroptosis etc. have been reported as possible causes of cancer cell elimination further corroborating existence of an alternative mechanism of drug activity. There are several reports which suggest autophagy as possible effecter of cell death [140,141]. Obatoclax can induce apoptosis in Beclin 1 dependent and independent manner [142,143,144,145]. There is evidence that suggests autophagy and apoptosis work in tandem to eliminate cancer cells [146]; however, there are contradictory reports that suggest obatoclax inhibits autophagy by impairing lysosomal function to induce cytotoxic effects in cancer [147,148]. In a recent study, Basit et al. reported necroptosis mediated cell death by induction of fusion of necrosome on autophagosomal membrane on treatment with obatoclax [149]. Champa et al. observed necrotic cell death in highly resistant anaplastic thyroid cancer cells upon treatment with OBT. On treatment OBT quickly localized in lysosomes and neutralized their pH. Interestingly, OBT activity was dependent on its accumulation in lysosome rather than its interaction with Bcl2 family proteins. OBT has demonstrated a potentiating effect on cancers in combination with other chemotherapeutics [150,151]. Li et al. reported a synergistic effect between OBT and cisplatin in non-small-cell lung cancer [152]. Resistance to therapy is a major concern in cancer treatment. OBT has shown great response in resistant cancer cells and has overcome chemoresistance in different cancers [153,154].
OBT has shown significant anticancer activity in leukemia cells which rely on Bcl2 family proteins for their survival. OBT has demonstrated specificity towards leukemia cancer stem cells [14]. Several studies have identified OBT as potential chemotherapeutic agent to overcome glucocorticoid resistance in leukemia [155,156]. Wei et al. revealed synergistic antileukemia activity of OBT with a histone deacetylase inhibitor [146]. OBT’s promising preclinical efficacy has not translated in clinical trials. Phase I studies of OBT in chronic lymphocytic leukemia (CLL) patients demonstrated significant toxicities such as somnolence, ataxia, and confusion with limited efficacy [157].
It is indisputable that OBT is a BH3 inhibitor; however, there is ample evidence that suggest an alternate mechanistic intervention for OBT’s anticancer activity. Furthermore, several studies indicated neutralization or alkalization of lysosomes upon treatment with OBT as the leading event in OBT induced cancer cell death suggesting ionophoretic activity of OBT playing a prominent role in anticancer activity of the drug. Also, OBT’s specific activity towards CSCs and its ability to sensitize resistant cancers indicates immense anticancer potential of OBT. It is imperative to understand various molecular targets and pathways involved in OBT induced therapeutic effects. OBT can be a lucrative therapeutic intervention for cancer sans its toxic side effects and perhaps a better understanding of its activity will render it more clinical relevance.

4. Ionophores in Ongoing Clinical Trials for Cancer Treatment

There are a few animal studies demonstrating that NIG, at tolerated doses, in combination with other compounds has antitumor effects [158]. However, at present there are no registered clinical trials using NIG as anticancer agents www.clinicaltrials.gov.
There are a few clinical reports in the literature about the use of SAL that comes from pilot studies involving a few patients (four metastatic breast cancer patients, a metastatic ovarian cancer patient, and a patient with head and neck squamous cell carcinoma) [9,159]. At present there are no registered clinical trials at www.clinicaltrials.gov.
OBT has been tested in multiple clinical trials as single agent as well as in combination with other anticancer agents (Table 2). As single agent OBT was not associated with an objective response in AML [160] or showed only a modest activity in heavily pretreated patients with advanced CLL [157], classic Hodgkin lymphoma (cHL) [161] myelodysplastic syndromes [162]. OBT in combination with carboplatin/etoposide failed to significantly improve objective response rate (ORR), progression-free survival (PFS) or overall survival (OS) in first-line treatment of extensive-stage small cell lung cancer (ES-SCLC) [163]. Similarly, when added to topotecan did not exceed the historic response rate seen with topotecan alone in patients with relapsed SCLC following the first-line platinum-based therapy [164].
There are several factors that may limit the clinical translation of ionophores as anticancer agent especially as single agents. On one hand, the pharmaceutical industry may be more interested in pursuing patentable drugs. However, toxicity may be the main concern. For instance, SAL is very toxic to other normal cells at concentrations effective against cancer stem-like cells [166] and therefore it is unlikely that this drug will be useful as single agent [167]. The ability of NIG to induce apoptosis or necrosis by increase K+ efflux occurs only at relatively high concentration (2.5–7.5 µM) [6] that may be not tolerated in vivo. However, as previously discussed, other biological effects at tolerated doses may be exploited for combination therapy in clinical trials.

5. Conclusions

The disruption of ion homeostasis important for proliferation and survival of cancer constitutes a potential target for chemotherapy. The ionophores SAL, NIG and OBT have shown important anticancer activities in in vitro and in vivo preclinical models of cancer as single agents as well as in combination with other anticancer drugs. More important, they also showed anticancer activity against putative cancer stem-like cells. The underlying reason some ionophores work against cancer stem cells and other ionophores do not is poorly understood but it is possible that other ionophore-independent activity target key processes associated with stemness, for instance, (i) SAL induces ER Ca2+ depletion up-regulating C/EBP homologous protein (CHOP), which inhibits Wnt signaling by down-regulating β-catenin [168]. SAL also inhibits K-ras [169], Notch [82] and Hedgehog signaling [170], (ii) NIG is also a potent modulator of the Wnt signaling pathway [6] and (iii) OBT targets cancer stem cells via disruption of BCL-2-dependent oxidative phosphorylation [14].
Except for OBT, these ionophores have not been translated into clinical trials. At present, the results of clinical trials with OBT as single agent or in combination with other anticancer drugs did not show a significant benefit. It is possible that the high toxicity of ionophores towards non-cancer cells may be limiting their clinical use. The selectivity towards non-cancer cells can be investigated by using non-cancer cell lines from the same organ. For instance, the Beas-2B cell line consists of epithelial cells that were isolated from normal human bronchial epithelium obtained from autopsy of non-cancerous individuals and is sometimes used to compare to lung cancer cells [171]. To overcome this limitation, future development in targeted drug delivery may help to improve the ability of these promising compounds. The use of different types SAL loaded nanoparticles alone or in combination with other drugs showed improved efficacy compared to SAL alone in a variety of cancer cell types (Table 3). Alternatively, SAL, NIG and OBT may serve as lead compounds to develop derivatives more selective towards non-cancer cells. In this context, several derivatives of OBT [172,173] and SAL [120,121,174,175,176,177,178,179] have shown anticancer effects. For instance, derivatives with chemical modification of the allylic C20 hydroxyl of SAL, located at the C-ring, enhanced the activity over 5-fold against breast cancer cells compared to the native structure [121]. Derivatives of OBT were also found to be more potent against PLC5 hepatocellular carcinoma cells than the original compound [173]. At present the biological effects and selectivity, in particular the ability to deplete chemoresistant cells such as cancer stem-like cells need to be further investigated in more advanced preclinical (animal) models. In summary, targeted delivery and development of more potent and selective synthetic derivatives of concerned ionophores can facilitate the translation into clinical applications for cancer treatment.

Author Contributions

V.K., J.S.Y., A.K., N.A. and A.K.V.I. contributed to researching data and writing the manuscript. V.K. conceived and directed the study.

Funding

Research in our lab is supported by NIH grants to N.A. and A.K.V.I.

Conflicts of Interest

The authors have declared no conflict of interest.

References

  1. Govorunova, E.G.; Sineshchekov, O.A.; Janz, R.; Liu, X.; Spudich, J.L. Natural light-gated anion channels: A family of microbial rhodopsins for advanced optogenetics. Science 2015, 349, 647–650. [Google Scholar] [CrossRef] [PubMed]
  2. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. Carrier Proteins and Active Membrane Transport. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002. [Google Scholar]
  3. Manago, A.; Leanza, L.; Carraretto, L.; Sassi, N.; Grancara, S.; Quintana-Cabrera, R.; Trimarco, V.; Toninello, A.; Scorrano, L.; Trentin, L.; et al. Early effects of the antineoplastic agent salinomycin on mitochondrial function. Cell Death Dis. 2015, 6, e1930. [Google Scholar] [CrossRef] [PubMed]
  4. Boesch, M.; Zeimet, A.G.; Rumpold, H.; Gastl, G.; Sopper, S.; Wolf, D. Drug Transporter-Mediated Protection of Cancer Stem Cells from Ionophore Antibiotics. Stem Cells Transl. Med. 2015, 4, 1028–1032. [Google Scholar] [CrossRef] [PubMed]
  5. 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]
  6. Yakisich, J.S.; Azad, N.; Kaushik, V.; O’Doherty, G.A.; Iyer, A.K. Nigericin decreases the viability of multidrug-resistant cancer cells and lung tumorspheres and potentiates the effects of cardiac glycosides. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. 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] [PubMed]
  8. Magrath, J.W.; Kim, Y. Salinomycin’s potential to eliminate glioblastoma stem cells and treat glioblastoma multiforme (Review). Int. J. Oncol. 2017, 51, 753–759. [Google Scholar] [CrossRef] [PubMed]
  9. Naujokat, C.; Steinhart, R. Salinomycin as a drug for targeting human cancer stem cells. J. Biomed. Biotechnol 2012, 2012, 950658. [Google Scholar] [CrossRef] [PubMed]
  10. Berghauser Pont, L.M.; Spoor, J.K.; Venkatesan, S.; Swagemakers, S.; Kloezeman, J.J.; Dirven, C.M.; van der Spek, P.J.; Lamfers, M.L.; Leenstra, S. The Bcl-2 inhibitor Obatoclax overcomes resistance to histone deacetylase inhibitors SAHA and LBH589 as radiosensitizers in patient-derived glioblastoma stem-like cells. Genes Cancer 2014, 5, 445–459. [Google Scholar] [PubMed]
  11. Blatt, K.; Herrmann, H.; Stefanzl, G.; Sperr, W.R.; Valent, P. Evaluation of in vitro effects of various targeted drugs on plasma cells and putative neoplastic stem cells in patients with multiple myeloma. Oncotarget 2016, 7, 65627–65642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cruickshanks, N.; Hamed, H.A.; Booth, L.; Tavallai, S.; Syed, J.; Sajithlal, G.B.; Grant, S.; Poklepovic, A.; Dent, P. Histone deacetylase inhibitors restore toxic BH3 domain protein expression in anoikis-resistant mammary and brain cancer stem cells, thereby enhancing the response to anti-ERBB1/ERBB2 therapy. Cancer Biol. Ther. 2013, 14, 982–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ishida, C.T.; Bianchetti, E.; Shu, C.; Halatsch, M.E.; Westhoff, M.A.; Karpel-Massler, G.; Siegelin, M.D. BH3-mimetics and BET-inhibitors elicit enhanced lethality in malignant glioma. Oncotarget 2017, 8, 29558–29573. [Google Scholar] [CrossRef] [PubMed]
  14. Lagadinou, E.D.; Sach, A.; Callahan, K.; Rossi, R.M.; Neering, S.J.; Minhajuddin, M.; Ashton, J.M.; Pei, S.; Grose, V.; O’Dwyer, K.M.; et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 2013, 12, 329–341. [Google Scholar] [CrossRef] [PubMed]
  15. Singh, S.; Bora-Singhal, N.; Kroeger, J.; Laklai, H.; Chellappan, S.P. βArrestin-1 and Mcl-1 modulate self-renewal growth of cancer stem-like side-population cells in non-small cell lung cancer. PLoS ONE 2013, 8, e55982. [Google Scholar] [CrossRef] [PubMed]
  16. David, J.M.; Owens, T.A.; Inge, L.J.; Bremner, R.M.; Rajasekaran, A.K. Gramicidin A blocks tumor growth and angiogenesis through inhibition of hypoxia-inducible factor in renal cell carcinoma. Mol. Cancer Ther. 2014, 13, 788–799. [Google Scholar] [CrossRef] [PubMed]
  17. Rao, D.K.; Liu, H.; Ambudkar, S.V.; Mayer, M. A combination of curcumin with either gramicidin or ouabain selectively kills cells that express the multidrug resistance-linked ABCG2 transporter. J. Biol. Chem. 2014, 289, 31397–31410. [Google Scholar] [CrossRef] [PubMed]
  18. Han, S.; Tie, X.; Meng, L.; Wang, Y.; Wu, A. PMA and ionomycin induce glioblastoma cell death: Activation-induced cell-death-like phenomena occur in glioma cells. PLoS ONE 2013, 8, e76717. [Google Scholar] [CrossRef] [PubMed]
  19. Miyake, H.; Hara, I.; Yamanaka, K.; Arakawa, S.; Kamidono, S. Calcium ionophore, ionomycin inhibits growth of human bladder cancer cells both in vitro and in vivo with alteration of Bcl-2 and Bax expression levels. J. Urol 1999, 162, 916–921. [Google Scholar] [CrossRef] [PubMed]
  20. Deng, Y.; Zhang, J.; Wang, Z.; Yan, Z.; Qiao, M.; Ye, J.; Wei, Q.; Wang, J.; Wang, X.; Zhao, L.; et al. Antibiotic monensin synergizes with EGFR inhibitors and oxaliplatin to suppress the proliferation of human ovarian cancer cells. Sci. Rep. 2015, 5, 17523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Kim, S.H.; Kim, K.Y.; Yu, S.N.; Park, S.G.; Yu, H.S.; Seo, Y.K.; Ahn, S.C. Monensin Induces PC-3 Prostate Cancer Cell Apoptosis via ROS Production and Ca2+ Homeostasis Disruption. Anticancer Res. 2016, 36, 5835–5843. [Google Scholar] [CrossRef] [PubMed]
  22. Tumova, L.; Pombinho, A.R.; Vojtechova, M.; Stancikova, J.; Gradl, D.; Krausova, M.; Sloncova, E.; Horazna, M.; Kriz, V.; Machonova, O.; et al. Monensin inhibits canonical Wnt signaling in human colorectal cancer cells and suppresses tumor growth in multiple intestinal neoplasia mice. Mol. Cancer Ther. 2014, 13, 812–822. [Google Scholar] [CrossRef] [PubMed]
  23. Ryoo, I.J.; Park, H.R.; Choo, S.J.; Hwang, J.H.; Park, Y.M.; Bae, K.H.; Shin-Ya, K.; Yoo, I.D. Selective cytotoxic activity of valinomycin against HT-29 Human colon carcinoma cells via down-regulation of GRP78. Biol. Pharm. Bull. 2006, 29, 817–820. [Google Scholar] [CrossRef] [PubMed]
  24. Daoud, S.S.; Forde, N.H. Synergistic cytotoxic actions of cisplatin and liposomal valinomycin on human ovarian carcinoma cells. Cancer Chemother. Pharmacol. 1991, 28, 370–376. [Google Scholar] [CrossRef] [PubMed]
  25. Kim, K.Y.; Kim, S.H.; Yu, S.N.; Park, S.G.; Kim, Y.W.; Nam, H.W.; An, H.H.; Yu, H.S.; Kim, Y.W.; Ji, J.H.; et al. Lasalocid induces cytotoxic apoptosis and cytoprotective autophagy through reactive oxygen species in human prostate cancer PC-3 cells. Biomed. Pharmacother. 2017, 88, 1016–1024. [Google Scholar] [CrossRef] [PubMed]
  26. Dornetshuber, R.; Heffeter, P.; Kamyar, M.R.; Peterbauer, T.; Berger, W.; Lemmens-Gruber, R. Enniatin exerts p53-dependent cytostatic and p53-independent cytotoxic activities against human cancer cells. Chem. Res. Toxicol. 2007, 20, 465–473. [Google Scholar] [CrossRef] [PubMed]
  27. Dornetshuber-Fleiss, R.; Heilos, D.; Mohr, T.; Richter, L.; Sussmuth, R.D.; Zlesak, M.; Novicky, A.; Heffeter, P.; Lemmens-Gruber, R.; Berger, W. The naturally born fusariotoxin enniatin B and sorafenib exert synergistic activity against cervical cancer in vitro and in vivo. Biochem. Pharmacol. 2015, 93, 318–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lu, C.L.; Lin, H.I.; Chen, B.F.; Jow, G.M. Beauvericin-induced cell apoptosis through the mitogen-activated protein kinase pathway in human nonsmall cell lung cancer A549 cells. J. Toxicol. Sci. 2016, 41, 429–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Watjen, W.; Debbab, A.; Hohlfeld, A.; Chovolou, Y.; Proksch, P. The mycotoxin beauvericin induces apoptotic cell death in H4IIE hepatoma cells accompanied by an inhibition of NF-κB-activity and modulation of MAP-kinases. Toxicol. Lett. 2014, 231, 9–16. [Google Scholar] [CrossRef] [PubMed]
  30. Prevarskaya, N.; Skryma, R.; Shuba, Y. Ion channels and the hallmarks of cancer. Trends Mol. Med. 2010, 16, 107–121. [Google Scholar] [CrossRef] [PubMed]
  31. Roderick, H.L.; Cook, S.J. Ca2+ signalling checkpoints in cancer: Remodelling Ca2+ for cancer cell proliferation and survival. Nat. Rev. Cancer 2008, 8, 361–375. [Google Scholar] [CrossRef] [PubMed]
  32. Kahl, C.R.; Means, A.R. Regulation of cell cycle progression by calcium/calmodulin-dependent pathways. Endocr. Rev. 2003, 24, 719–736. [Google Scholar] [CrossRef] [PubMed]
  33. Thebault, S.; Flourakis, M.; Vanoverberghe, K.; Vandermoere, F.; Roudbaraki, M.; Lehen’kyi, V.; Slomianny, C.; Beck, B.; Mariot, P.; Bonnal, J.L.; et al. Differential role of transient receptor potential channels in Ca2+ entry and proliferation of prostate cancer epithelial cells. Cancer Res. 2006, 66, 2038–2047. [Google Scholar] [CrossRef] [PubMed]
  34. Wei, C.; Wang, X.; Chen, M.; Ouyang, K.; Zheng, M.; Cheng, H. Flickering calcium microdomains signal turning of migrating cells. Can. J. Physiol. Pharmacol. 2010, 88, 105–110. [Google Scholar] [CrossRef] [PubMed]
  35. Deliot, N.; Constantin, B. Plasma membrane calcium channels in cancer: Alterations and consequences for cell proliferation and migration. Biochim. Biophys. Acta 2015, 1848 Pt B, 2512–2522. [Google Scholar] [CrossRef]
  36. Dubois, C.; Vanden Abeele, F.; Prevarskaya, N. Targeting apoptosis by the remodelling of calcium-transporting proteins in cancerogenesis. FEBS J. 2013, 280, 5500–5510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of cell death: The calcium-apoptosis link. Nat. Rev. Mol. Cell Biol. 2003, 4, 552–565. [Google Scholar] [CrossRef] [PubMed]
  38. Hoffmann, E.K.; Lambert, I.H. Ion channels and transporters in the development of drug resistance in cancer cells. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130109. [Google Scholar] [CrossRef] [PubMed]
  39. Wang, Z. Roles of K+ channels in regulating tumour cell proliferation and apoptosis. Pflugers Arch. 2004, 448, 274–286. [Google Scholar] [CrossRef] [PubMed]
  40. Fraser, S.P.; Ozerlat-Gunduz, I.; Brackenbury, W.J.; Fitzgerald, E.M.; Campbell, T.M.; Coombes, R.C.; Djamgoz, M.B. Regulation of voltage-gated sodium channel expression in cancer: Hormones, growth factors and auto-regulation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130105. [Google Scholar] [CrossRef] [PubMed]
  41. Lopez, V.; Kelleher, S.L. Zip6-attenuation promotes epithelial-to-mesenchymal transition in ductal breast tumor (T47D) cells. Exp. Cell Res. 2010, 316, 366–375. [Google Scholar] [CrossRef] [PubMed]
  42. Matsui, C.; Takatani-Nakase, T.; Hatano, Y.; Kawahara, S.; Nakase, I.; Takahashi, K. Zinc and its transporter ZIP6 are key mediators of breast cancer cell survival under high glucose conditions. FEBS Lett. 2017, 591, 3348–3359. [Google Scholar] [CrossRef] [PubMed]
  43. Donadelli, M.; Dalla Pozza, E.; Scupoli, M.T.; Costanzo, C.; Scarpa, A.; Palmieri, M. Intracellular zinc increase inhibits p53(−/−) pancreatic adenocarcinoma cell growth by ROS/AIF-mediated apoptosis. Biochim. Biophys. Acta 2009, 1793, 273–280. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, Y.; Wei, X.; Yan, P.; Han, Y.; Sun, S.; Wu, K.; Fan, D. Human mitochondrial Mrs2 protein promotes multidrug resistance in gastric cancer cells by regulating p27, cyclin D1 expression and cytochrome C release. Cancer Biol. Ther. 2009, 8, 607–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Castiglioni, S.; Cazzaniga, A.; Trapani, V.; Cappadone, C.; Farruggia, G.; Merolle, L.; Wolf, F.I.; Iotti, S.; Maier, J.A. Magnesium homeostasis in colon carcinoma LoVo cells sensitive or resistant to doxorubicin. Sci. Rep. 2015, 5, 16538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Furukawa, T.; Komatsu, M.; Ikeda, R.; Tsujikawa, K.; Akiyama, S. Copper transport systems are involved in multidrug resistance and drug transport. Curr. Med. Chem. 2008, 15, 3268–3278. [Google Scholar] [CrossRef] [PubMed]
  47. Bompiani, K.M.; Tsai, C.Y.; Achatz, F.P.; Liebig, J.K.; Howell, S.B. Copper transporters and chaperones CTR1, CTR2, ATOX1, and CCS as determinants of cisplatin sensitivity. Metallomics 2016, 8, 951–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Lee, Y.Y.; Choi, C.H.; Do, I.G.; Song, S.Y.; Lee, W.; Park, H.S.; Song, T.J.; Kim, M.K.; Kim, T.J.; Lee, J.W.; et al. Prognostic value of the copper transporters, CTR1 and CTR2, in patients with ovarian carcinoma receiving platinum-based chemotherapy. Gynecol. Oncol. 2011, 122, 361–365. [Google Scholar] [CrossRef] [PubMed]
  49. Nakagawa, T.; Inoue, Y.; Kodama, H.; Yamazaki, H.; Kawai, K.; Suemizu, H.; Masuda, R.; Iwazaki, M.; Yamada, S.; Ueyama, Y.; et al. Expression of copper-transporting p-type adenosine triphosphatase (ATP7B) correlates with cisplatin resistance in human non-small cell lung cancer xenografts. Oncol. Rep. 2008, 20, 265–270. [Google Scholar] [PubMed]
  50. Samimi, G.; Safaei, R.; Katano, K.; Holzer, A.K.; Rochdi, M.; Tomioka, M.; Goodman, M.; Howell, S.B. Increased expression of the copper efflux transporter ATP7A mediates resistance to cisplatin, carboplatin, and oxaliplatin in ovarian cancer cells. Clin. Cancer Res. 2004, 10, 4661–4669. [Google Scholar] [CrossRef] [PubMed]
  51. Ober, S.S.; Pardee, A.B. Intracellular pH is increased after transformation of Chinese hamster embryo fibroblasts. Proc. Natl. Acad. Sci. USA 1987, 84, 2766–2770. [Google Scholar] [CrossRef] [PubMed]
  52. Larsen, A.K.; Escargueil, A.E.; Skladanowski, A. Resistance mechanisms associated with altered intracellular distribution of anticancer agents. Pharmacol. Ther. 2000, 85, 217–229. [Google Scholar] [CrossRef]
  53. Cardone, R.A.; Casavola, V.; Reshkin, S.J. The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis. Nat. Rev. Cancer 2005, 5, 786–795. [Google Scholar] [CrossRef] [PubMed]
  54. Abdoul-Azize, S.; Dubus, I.; Vannier, J.P. Improvement of dexamethasone sensitivity by chelation of intracellular Ca2+ in pediatric acute lymphoblastic leukemia cells through the prosurvival kinase ERK1/2 deactivation. Oncotarget 2017, 8, 27339–27352. [Google Scholar] [CrossRef] [PubMed]
  55. Hsia, T.C.; Huang, Y.P.; Jiang, Y.W.; Chen, H.Y.; Cheng, Z.Y.; Hsiao, Y.T.; Chen, C.Y.; Peng, S.F.; Chueh, F.S.; Chou, Y.C.; et al. Phenethyl Isothiocyanate Induces Apoptotic Cell Death Through the Mitochondria-dependent Pathway in Gefitinib-resistant NCI-H460 Human Lung Cancer Cells In Vitro. Anticancer Res. 2018, 38, 2137–2147. [Google Scholar] [PubMed]
  56. Chen, S.J.; Kuo, C.C.; Pan, H.Y.; Tsou, T.C.; Yeh, S.C.; Chang, J.Y. Desferal regulates hCtr1 and transferrin receptor expression through Sp1 and exhibits synergistic cytotoxicity with platinum drugs in oxaliplatin-resistant human cervical cancer cells in vitro and in vivo. Oncotarget 2016, 7, 49310–49321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Lee, E.L.; Shimizu, T.; Ise, T.; Numata, T.; Kohno, K.; Okada, Y. Impaired activity of volume-sensitive Cl-channel is involved in cisplatin resistance of cancer cells. J. Cell. Physiol. 2007, 211, 513–521. [Google Scholar] [CrossRef] [PubMed]
  58. Goh, W.; Sleptsova-Freidrich, I.; Petrovic, N. Use of proton pump inhibitors as adjunct treatment for triple-negative breast cancers. An introductory study. J. Pharm. Pharm. Sci. 2014, 17, 439–446. [Google Scholar] [CrossRef] [PubMed]
  59. Kaushik, V.; Yakisich, J.S.; Azad, N.; Kulkarni, Y.; Venkatadri, R.; Wright, C.; Rojanasakul, Y.; Iyer, A.K.V. Anti-Tumor Effects of Cardiac Glycosides on Human Lung Cancer Cells and Lung Tumorspheres. J. Cell. Physiol. 2017, 232, 2497–2507. [Google Scholar] [CrossRef] [PubMed]
  60. Kaushik, V.; Azad, N.; Yakisich, J.S.; Iyer, A.K. Antitumor effects of naturally occurring cardiac glycosides convallatoxin and peruvoside on human ER+ and triple-negative breast cancers. Cell Death Discov. 2017, 3, 17009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Kulkarni, Y.M.; Kaushik, V.; Azad, N.; Wright, C.; Rojanasakul, Y.; O’Doherty, G.; Iyer, A.K. Autophagy-Induced Apoptosis in Lung Cancer Cells by a Novel Digitoxin Analog. J. Cell. Physiol. 2016, 231, 817–828. [Google Scholar] [CrossRef] [PubMed]
  62. Kulkarni, Y.M.; Yakisich, J.S.; Azad, N.; Venkatadri, R.; Kaushik, V.; O’Doherty, G.; Iyer, A.K.V. Anti-tumorigenic effects of a novel digitoxin derivative on both estrogen receptor-positive and triple-negative breast cancer cells. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [PubMed]
  63. Babcock, J.J.; Li, M. hERG channel function: Beyond long QT. Acta Pharmacol. Sin. 2013, 34, 329–335. [Google Scholar] [CrossRef] [PubMed]
  64. Pillozzi, S.; Masselli, M.; De Lorenzo, E.; Accordi, B.; Cilia, E.; Crociani, O.; Amedei, A.; Veltroni, M.; D’Amico, M.; Basso, G.; et al. Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers. Blood 2011, 117, 902–914. [Google Scholar] [CrossRef] [PubMed]
  65. Arcangeli, A.; Becchetti, A. Novel perspectives in cancer therapy: Targeting ion channels. Drug Resist. Updat. 2015, 21–22, 11–19. [Google Scholar] [CrossRef] [PubMed]
  66. Yakisich, J.S.; Kulkarni, Y.; Azad, N.; Iyer, A.K.V. Selective and Irreversible Induction of Necroptotic Cell Death in Lung Tumorspheres by Short-Term Exposure to Verapamil in Combination with Sorafenib. Stem Cells Int. 2017, 2017, 5987015. [Google Scholar] [CrossRef] [PubMed]
  67. Helson, L. Calcium channel blocker enhancement of anticancer drug cytotoxicity—A review. Cancer Drug Deliv. 1984, 1, 353–361. [Google Scholar] [CrossRef] [PubMed]
  68. Vilpo, J.; Koski, T.; Vilpo, L. Calcium antagonists potentiate p-glycoprotein-independent anticancer drugs in chronic lymphocytic leukemia cells in vitro. Haematologica 2000, 85, 806–813. [Google Scholar] [PubMed]
  69. Hilgenfeld, R.; Saenger, W. Structural chemistry of natural and synthetic ionophores and their complexes with cations. Top. Curr. Chem. 1982, 101, 1–82. [Google Scholar] [PubMed]
  70. Antonenko, Y.N.; Rokitskaya, T.I.; Huczynski, A. Electrogenic and nonelectrogenic ion fluxes across lipid and mitochondrial membranes mediated by monensin and monensin ethyl ester. Biochim. Biophys. Acta 2015, 1848, 995–1004. [Google Scholar] [CrossRef] [PubMed]
  71. Huczynski, A. Polyether ionophores-promising bioactive molecules for cancer therapy. Bioorg. Med. Chem. Lett. 2012, 22, 7002–7010. [Google Scholar] [CrossRef] [PubMed]
  72. Versini, A.; Saier, L.; Sindikubwabo, F.; Müller, S.; Cañeque, T.; Rodriguez, R. Chemical biology of salinomycin. Rodriguez 2018, 74, 5585–5614. [Google Scholar] [CrossRef]
  73. Rutkowski, J.; Brzezinski, B. Structures and properties of naturally occurring polyether antibiotics. Biomed. Res. Int. 2013, 2013, 162513. [Google Scholar] [CrossRef] [PubMed]
  74. An, H.; Kim, J.Y.; Lee, N.; Cho, Y.; Oh, E.; Seo, J.H. Salinomycin possesses anti-tumor activity and inhibits breast cancer stem-like cells via an apoptosis-independent pathway. Biochem. Biophys. Res. Commun. 2015, 466, 696–703. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, T.; Yi, L.; Li, F.; Hu, R.; Hu, S.; Yin, Y.; Lan, C.; Li, Z.; Fu, C.; Cao, L.; et al. Salinomycin inhibits the tumor growth of glioma stem cells by selectively suppressing glioma-initiating cells. Mol. Med. Rep. 2015, 11, 2407–2412. [Google Scholar] [CrossRef] [PubMed]
  76. Chung, H.; Kim, Y.H.; Kwon, M.; Shin, S.J.; Kwon, S.H.; Cha, S.D.; Cho, C.H. The effect of salinomycin on ovarian cancer stem-like cells. Obstet. Gynecol. Sci. 2016, 59, 261–268. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, H.G.; Shin, S.J.; Chung, H.W.; Kwon, S.H.; Cha, S.D.; Lee, J.E.; Cho, C.H. Salinomycin reduces stemness and induces apoptosis on human ovarian cancer stem cell. J. Gynecol. Oncol. 2017, 28, e14. [Google Scholar] [CrossRef] [PubMed]
  78. Mao, J.; Fan, S.; Ma, W.; Fan, P.; Wang, B.; Zhang, J.; Wang, H.; Tang, B.; Zhang, Q.; Yu, X.; et al. Roles of Wnt/beta-catenin signaling in the gastric cancer stem cells proliferation and salinomycin treatment. Cell Death Dis. 2014, 5, e1039. [Google Scholar] [CrossRef] [PubMed]
  79. Tang, Q.L.; Zhao, Z.Q.; Li, J.C.; Liang, Y.; Yin, J.Q.; Zou, C.Y.; Xie, X.B.; Zeng, Y.X.; Shen, J.N.; Kang, T.; et al. Salinomycin inhibits osteosarcoma by targeting its tumor stem cells. Cancer Lett. 2011, 311, 113–121. [Google Scholar] [CrossRef] [PubMed]
  80. Zhang, C.; Tian, Y.; Song, F.; Fu, C.; Han, B.; Wang, Y. Salinomycin inhibits the growth of colorectal carcinoma by targeting tumor stem cells. Oncol. Rep. 2015, 34, 2469–2476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Antoszczak, M.; Huczynski, A. Anticancer Activity of Polyether Ionophore-Salinomycin. Anticancer Agents Med. Chem. 2015, 15, 575–591. [Google Scholar] [CrossRef] [PubMed]
  82. Dewangan, J.; Srivastava, S.; Rath, S.K. Salinomycin: A new paradigm in cancer therapy. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kaplan, F.; Teksen, F. Apoptotic effects of salinomycin on human ovarian cancer cell line (OVCAR-3). Tumour Biol. 2016, 37, 3897–3903. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, K.Y.; Yu, S.N.; Lee, S.Y.; Chun, S.S.; Choi, Y.L.; Park, Y.M.; Song, C.S.; Chatterjee, B.; Ahn, S.C. Salinomycin-induced apoptosis of human prostate cancer cells due to accumulated reactive oxygen species and mitochondrial membrane depolarization. Biochem. Biophys. Res. Commun. 2011, 413, 80–86. [Google Scholar] [CrossRef] [PubMed]
  85. Lee, H.G.; Lee, J.M.; Shin, S.J.; Kwon, S.H.; Lee, G.S.; Song, C.H.; Choi, E.S.; Cha, S.D.; Cho, C.H. Salinomycin inhibited cell proliferation and induced apoptosis in human uterine leiomyoma cells. Obstet. Gynecol. Sci. 2014, 57, 501–506. [Google Scholar] [CrossRef] [PubMed]
  86. Lu, D.; Choi, M.Y.; Yu, J.; Castro, J.E.; Kipps, T.J.; Carson, D.A. Salinomycin inhibits Wnt signaling and selectively induces apoptosis in chronic lymphocytic leukemia cells. Proc. Natl. Acad. Sci. USA 2011, 108, 13253–13257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wang, F.; He, L.; Dai, W.Q.; Xu, Y.P.; Wu, D.; Lin, C.L.; Wu, S.M.; Cheng, P.; Zhang, Y.; Shen, M.; et al. Salinomycin inhibits proliferation and induces apoptosis of human hepatocellular carcinoma cells in vitro and in vivo. PLoS ONE 2012, 7, e50638. [Google Scholar] [CrossRef] [PubMed]
  88. Wu, D.; Zhang, Y.; Huang, J.; Fan, Z.; Shi, F.; Wang, S. Salinomycin inhibits proliferation and induces apoptosis of human nasopharyngeal carcinoma cell in vitro and suppresses tumor growth in vivo. Biochem. Biophys. Res. Commun. 2014, 443, 712–717. [Google Scholar] [CrossRef] [PubMed]
  89. Li, T.; Su, L.; Zhong, N.; Hao, X.; Zhong, D.; Singhal, S.; Liu, X. Salinomycin induces cell death with autophagy through activation of endoplasmic reticulum stress in human cancer cells. Autophagy 2013, 9, 1057–1068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Verdoodt, B.; Vogt, M.; Schmitz, I.; Liffers, S.T.; Tannapfel, A.; Mirmohammadsadegh, A. Salinomycin induces autophagy in colon and breast cancer cells with concomitant generation of reactive oxygen species. PLoS ONE 2012, 7, e44132. [Google Scholar] [CrossRef] [PubMed]
  91. Endo, S.; Nakata, K.; Sagara, A.; Koikawa, K.; Ando, Y.; Kibe, S.; Takesue, S.; Nakayama, H.; Abe, T.; Okumura, T.; et al. Autophagy inhibition enhances antiproliferative effect of salinomycin in pancreatic cancer cells. Pancreatology 2017, 17, 990–996. [Google Scholar] [CrossRef] [PubMed]
  92. Kim, K.Y.; Park, K.I.; Kim, S.H.; Yu, S.N.; Park, S.G.; Kim, Y.W.; Seo, Y.K.; Ma, J.Y.; Ahn, S.C. Inhibition of Autophagy Promotes Salinomycin-Induced Apoptosis via Reactive Oxygen Species-Mediated PI3K/AKT/mTOR and ERK/p38 MAPK-Dependent Signaling in Human Prostate Cancer Cells. Int. J. Mol. Sci. 2017, 18, 1088. [Google Scholar] [CrossRef] [PubMed]
  93. Klose, J.; Stankov, M.V.; Kleine, M.; Ramackers, W.; Panayotova-Dimitrova, D.; Jager, M.D.; Klempnauer, J.; Winkler, M.; Bektas, H.; Behrens, G.M.; et al. Inhibition of autophagic flux by salinomycin results in anti-cancer effect in hepatocellular carcinoma cells. PLoS ONE 2014, 9, e95970. [Google Scholar] [CrossRef] [PubMed]
  94. Xipell, E.; Gonzalez-Huarriz, M.; Martinez de Irujo, J.J.; Garcia-Garzon, A.; Lang, F.F.; Jiang, H.; Fueyo, J.; Gomez-Manzano, C.; Alonso, M.M. Salinomycin induced ROS results in abortive autophagy and leads to regulated necrosis in glioblastoma. Oncotarget 2016, 7, 30626–30641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yue, W.; Hamai, A.; Tonelli, G.; Bauvy, C.; Nicolas, V.; Tharinger, H.; Codogno, P.; Mehrpour, M. Inhibition of the autophagic flux by salinomycin in breast cancer stem-like/progenitor cells interferes with their maintenance. Autophagy 2013, 9, 714–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kim, K.Y.; Park, K.I.; Kim, S.H.; Yu, S.N.; Lee, D.; Kim, Y.W.; Noh, K.T.; Ma, J.Y.; Seo, Y.K.; Ahn, S.C. Salinomycin Induces Reactive Oxygen Species and Apoptosis in Aggressive Breast Cancer Cells as Mediated with Regulation of Autophagy. Anticancer Res. 2017, 37, 1747–1758. [Google Scholar] [PubMed]
  97. Kim, S.H.; Choi, Y.J.; Kim, K.Y.; Yu, S.N.; Seo, Y.K.; Chun, S.S.; Noh, K.T.; Suh, J.T.; Ahn, S.C. Salinomycin simultaneously induces apoptosis and autophagy through generation of reactive oxygen species in osteosarcoma U2OS cells. Biochem. Biophys. Res. Commun. 2016, 473, 607–613. [Google Scholar] [CrossRef] [PubMed]
  98. Qin, L.S.; Jia, P.F.; Zhang, Z.Q.; Zhang, S.M. ROS-p53-cyclophilin-D signaling mediates salinomycin-induced glioma cell necrosis. J. Exp. Clin. Cancer Res. 2015, 34, 57. [Google Scholar] [CrossRef] [PubMed]
  99. He, L.; Wang, F.; Dai, W.Q.; Wu, D.; Lin, C.L.; Wu, S.M.; Cheng, P.; Zhang, Y.; Shen, M.; Wang, C.F.; et al. Mechanism of action of salinomycin on growth and migration in pancreatic cancer cell lines. Pancreatology 2013, 13, 72–78. [Google Scholar] [CrossRef] [PubMed]
  100. Klose, J.; Eissele, J.; Volz, C.; Schmitt, S.; Ritter, A.; Ying, S.; Schmidt, T.; Heger, U.; Schneider, M.; Ulrich, A. Salinomycin inhibits metastatic colorectal cancer growth and interferes with Wnt/beta-catenin signaling in CD133+ human colorectal cancer cells. BMC Cancer 2016, 16, 896. [Google Scholar] [CrossRef] [PubMed]
  101. Li, R.; Dong, T.; Hu, C.; Lu, J.; Dai, J.; Liu, P. Salinomycin repressed the epithelial-mesenchymal transition of epithelial ovarian cancer cells via downregulating Wnt/beta-catenin pathway. Onco Targets Ther. 2017, 10, 1317–1325. [Google Scholar] [CrossRef] [PubMed]
  102. Lu, W.; Li, Y. Salinomycin suppresses LRP6 expression and inhibits both Wnt/beta-catenin and mTORC1 signaling in breast and prostate cancer cells. J. Cell. Biochem. 2014, 115, 1799–1807. [Google Scholar] [CrossRef] [PubMed]
  103. 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] [PubMed]
  104. 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 2015, 44, 395–398. [Google Scholar] [PubMed]
  105. Kim, K.Y.; Kim, S.H.; Yu, S.N.; Park, S.K.; Choi, H.D.; Yu, H.S.; Ji, J.H.; Seo, Y.K.; Ahn, S.C. Salinomycin enhances doxorubicin-induced cytotoxicity in multidrug resistant MCF-7/MDR human breast cancer cells via decreased efflux of doxorubicin. Mol. Med. Rep. 2015, 12, 1898–1904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Oak, P.S.; Kopp, F.; Thakur, C.; Ellwart, J.W.; Rapp, U.R.; Ullrich, A.; Wagner, E.; Knyazev, P.; Roidl, A. Combinatorial treatment of mammospheres with trastuzumab and salinomycin efficiently targets HER2-positive cancer cells and cancer stem cells. Int. J. Cancer 2012, 131, 2808–2819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Sommer, A.K.; Hermawan, A.; Mickler, F.M.; Ljepoja, B.; Knyazev, P.; Brauchle, C.; Ullrich, A.; Wagner, E.; Roidl, A. Salinomycin co-treatment enhances tamoxifen cytotoxicity in luminal A breast tumor cells by facilitating lysosomal degradation of receptor tyrosine kinases. Oncotarget 2016, 7, 50461–50476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Zhang, G.N.; Liang, Y.; Zhou, L.J.; Chen, S.P.; Chen, G.; Zhang, T.P.; Kang, T.; Zhao, Y.P. Combination of salinomycin and gemcitabine eliminates pancreatic cancer cells. Cancer Lett. 2011, 313, 137–144. [Google Scholar] [CrossRef] [PubMed]
  109. 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] [PubMed]
  110. Manmuan, S.; Sakunrangsit, N.; Ketchart, W. Salinomycin overcomes acquired tamoxifen resistance through AIB1 and inhibits cancer cell invasion in endocrine resistant breast cancer. Clin. Exp. Pharmacol. Physiol. 2017, 44, 1042–1052. [Google Scholar] [CrossRef] [PubMed]
  111. Zhou, J.; Li, P.; Xue, X.; He, S.; Kuang, Y.; Zhao, H.; Chen, S.; Zhi, Q.; Guo, X. Salinomycin induces apoptosis in cisplatin-resistant colorectal cancer cells by accumulation of reactive oxygen species. Toxicol. Lett. 2013, 222, 139–145. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, Y.; Liang, C.; Xue, F.; Chen, W.; Zhi, X.; Feng, X.; Bai, X.; Liang, T. Salinomycin decreases doxorubicin resistance in hepatocellular carcinoma cells by inhibiting the beta-catenin/TCF complex association via FOXO3a activation. Oncotarget 2015, 6, 10350–10365. [Google Scholar] [PubMed]
  113. Hermawan, A.; Wagner, E.; Roidl, A. Consecutive salinomycin treatment reduces doxorubicin resistance of breast tumor cells by diminishing drug efflux pump expression and activity. Oncol. Rep. 2016, 35, 1732–1740. [Google Scholar] [CrossRef] [PubMed]
  114. Riccioni, R.; Dupuis, M.L.; Bernabei, M.; Petrucci, E.; Pasquini, L.; Mariani, G.; Cianfriglia, M.; Testa, U. The cancer stem cell selective inhibitor salinomycin is a p-glycoprotein inhibitor. Blood Cells Mol. Dis. 2010, 45, 86–92. [Google Scholar] [CrossRef] [PubMed]
  115. Jiang, J.; Chen, H.; Yu, C.; Zhang, Y.; Chen, M.; Tian, S.; Sun, C. The promotion of salinomycin delivery to hepatocellular carcinoma cells through EGFR and CD133 aptamers conjugation by PLGA nanoparticles. Nanomedicine (Lond.) 2015, 10, 1863–1879. [Google Scholar] [CrossRef] [PubMed]
  116. Kim, Y.J.; Liu, Y.; Li, S.; Rohrs, J.; Zhang, R.; Zhang, X.; Wang, P. Co-Eradication of Breast Cancer Cells and Cancer Stem Cells by Cross-Linked Multilamellar Liposomes Enhances Tumor Treatment. Mol. Pharm. 2015, 12, 2811–2822. [Google Scholar] [CrossRef] [PubMed]
  117. Mao, X.; Liu, J.; Gong, Z.; Zhang, H.; Lu, Y.; Zou, H.; Yu, Y.; Chen, Y.; Sun, Z.; Li, W.; et al. iRGD-conjugated DSPE-PEG2000 nanomicelles for targeted delivery of salinomycin for treatment of both liver cancer cells and cancer stem cells. Nanomedicine (Lond.) 2015, 10, 2677–2695. [Google Scholar] [CrossRef] [PubMed]
  118. Ni, M.; Xiong, M.; Zhang, X.; Cai, G.; Chen, H.; Zeng, Q.; Yu, Z. Poly (lactic-co-glycolic acid) nanoparticles conjugated with CD133 aptamers for targeted salinomycin delivery to CD133+ osteosarcoma cancer stem cells. Int. J. Nanomed. 2015, 10, 2537–2554. [Google Scholar]
  119. Yao, H.J.; Zhang, Y.G.; Sun, L.; Liu, Y. The effect of hyaluronic acid functionalized carbon nanotubes loaded with salinomycin on gastric cancer stem cells. Biomaterials 2014, 35, 9208–9223. [Google Scholar] [CrossRef] [PubMed]
  120. Huang, M.; Deng, Z.; Tian, J.; Liu, T. Synthesis and biological evaluation of salinomycin triazole analogues as anticancer agents. Eur. J. Med. Chem. 2017, 127, 900–908. [Google Scholar] [CrossRef] [PubMed]
  121. Huang, X.; Borgstrom, B.; Mansson, L.; Persson, L.; Oredsson, S.; Hegardt, C.; Strand, D. Semisynthesis of SY-1 for investigation of breast cancer stem cell selectivity of C-ring-modified salinomycin analogues. ACS Chem. Biol. 2014, 9, 1587–1594. [Google Scholar] [CrossRef] [PubMed]
  122. Benedict, R.G. Antibiotics produced by actinomycetes. Bot. Rev. 1953, 19. [Google Scholar] [CrossRef]
  123. Harned, R.L.; Hidy, P.H.; Corum, C.J.; Jones, K.L. Nigericin a new crystalline antibiotic from an unidentified Streptomyces. Antibiot. Chemother. (Northfield) 1951, 1, 594–596. [Google Scholar] [PubMed]
  124. Pouyssegur, J.; Franchi, A.; L’Allemain, G.; Paris, S. Cytoplasmic pH, a key determinant of growth factor-induced DNA synthesis in quiescent fibroblasts. FEBS Lett. 1985, 190, 115–119. [Google Scholar] [CrossRef] [Green Version]
  125. Moolenaar, W.H. Effects of growth factors on intracellular pH regulation. Annu. Rev. Physiol. 1986, 48, 363–376. [Google Scholar] [CrossRef] [PubMed]
  126. Damaghi, M.; Wojtkowiak, J.W.; Gillies, R.J. pH sensing and regulation in cancer. Front. Physiol. 2013, 4, 370. [Google Scholar] [CrossRef] [PubMed]
  127. Margolis, L.B.; Rozovskaja, I.A.; Skulachev, V.P. Acidification of the interior of Ehrlich ascites tumor cells by nigericin inhibits DNA synthesis. FEBS Lett. 1987, 220, 288–290. [Google Scholar] [CrossRef] [Green Version]
  128. Hegazy, A.M.; Yamada, D.; Kobayashi, M.; Kohno, S.; Ueno, M.; Ali, M.A.; Ohta, K.; Tadokoro, Y.; Ino, Y.; Todo, T.; et al. Therapeutic Strategy for Targeting Aggressive Malignant Gliomas by Disrupting Their Energy Balance. J. Biol. Chem. 2016, 291, 21496–21509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Varnes, M.E.; Clay, M.E.; Freeman, K.; Antunez, A.R.; Oleinick, N.L. Enhancement of photodynamic cell killing (with chloroaluminum phthalocyanine) by treatment of V79 cells with the ionophore nigericin. Cancer Res. 1990, 50, 1620–1625. [Google Scholar] [PubMed]
  130. Xue, L.Y.; Agarwal, M.L.; Varnes, M.E. Elevation of GRP-78 and loss of HSP-70 following photodynamic treatment of V79 cells: Sensitization by nigericin. Photochem. Photobiol. 1995, 62, 135–143. [Google Scholar] [CrossRef] [PubMed]
  131. Varnes, M.E.; Bayne, M.T.; Menegay, H.J.; Tuttle, S.W. Effect of the K+/H+ ionophore nigericin on response of A549 cells to photodynamic therapy and tert-butylhydroperoxide. Free Radic. Biol. Med. 1993, 15, 395–405. [Google Scholar] [CrossRef]
  132. Yang, L.; Mei, Y.; Xie, Q.; Han, X.; Zhang, F.; Gu, L.; Zhang, Y.; Chen, Y.; Li, G.; Gao, Z. Acidification induces Bax translocation to the mitochondria and promotes ultraviolet light-induced apoptosis. Cell. Mol. Biol. Lett. 2008, 13, 119–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Varnes, M.E.; Menegay, H.J.; McKenna, D.S. Inhibition of recovery from potentially lethal radiation damage in A549 cells by the K+/H+ ionophore nigericin. Int. J. Radiat. Oncol. Biol. Phys. 1991, 20, 281–285. [Google Scholar] [CrossRef]
  134. Wood, P.J.; Sansom, J.M.; Newell, K.; Tannock, I.F.; Stratford, I.J. Reduction of tumour intracellular pH and enhancement of melphalan cytotoxicity by the ionophore Nigericin. Int. J. Cancer 1995, 60, 264–268. [Google Scholar] [CrossRef] [PubMed]
  135. Jahde, E.; Glusenkamp, K.H.; Rajewsky, M.F. Nigericin enhances mafosfamide cytotoxicity at low extracellular pH. Cancer Chemother. Pharmacol. 1991, 27, 440–444. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, W.; Zhao, Y.; Yao, S.; Cui, X.; Pan, W.; Huang, W.; Gao, J.; Dong, T.; Zhang, S. Nigericin Inhibits Epithelial Ovarian Cancer Metastasis by Suppressing the Cell Cycle and Epithelial-Mesenchymal Transition. Biochemistry 2017, 82, 933–941. [Google Scholar] [CrossRef] [PubMed]
  137. 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]
  138. Vu, H.T.; Kobayashi, M.; Hegazy, A.M.; Tadokoro, Y.; Ueno, M.; Kasahara, A.; Takase, Y.; Nomura, N.; Peng, H.; Ito, C.; et al. Autophagy inhibition synergizes with calcium mobilization to achieve efficient therapy of malignant gliomas. Cancer Sci. 2018, 109, 2497–2508. [Google Scholar] [CrossRef] [PubMed]
  139. Diaz de Grenu, B.; Iglesias Hernandez, P.; Espona, M.; Quinonero, D.; Light, M.E.; Torroba, T.; Perez-Tomas, R.; Quesada, R. Synthetic prodiginine obatoclax (GX15-070) and related analogues: Anion binding, transmembrane transport, and cytotoxicity properties. Chemistry 2011, 17, 14074–14083. [Google Scholar] [CrossRef] [PubMed]
  140. Cruickshanks, N.; Tang, Y.; Booth, L.; Hamed, H.; Grant, S.; Dent, P. Lapatinib and obatoclax kill breast cancer cells through reactive oxygen species-dependent endoplasmic reticulum stress. Mol. Pharmacol. 2012, 82, 1217–1229. [Google Scholar] [CrossRef] [PubMed]
  141. Martin, A.P.; Mitchell, C.; Rahmani, M.; Nephew, K.P.; Grant, S.; Dent, P. Inhibition of MCL-1 enhances lapatinib toxicity and overcomes lapatinib resistance via BAK-dependent autophagy. Cancer Biol. Ther. 2009, 8, 2084–2096. [Google Scholar] [CrossRef] [PubMed]
  142. Brem, E.A.; Thudium, K.; Khubchandani, S.; Tsai, P.C.; Olejniczak, S.H.; Bhat, S.; Riaz, W.; Gu, J.; Iqbal, A.; Campagna, R.; et al. Hernandez-Ilizaliturri, F.J. Distinct cellular and therapeutic effects of obatoclax in rituximab-sensitive and -resistant lymphomas. Br. J. Haematol. 2011, 153, 599–611. [Google Scholar] [CrossRef] [PubMed]
  143. Liang, L.Z.; Ma, B.; Liang, Y.J.; Liu, H.C.; Zhang, T.H.; Zheng, G.S.; Su, Y.X.; Liao, G.Q. Obatoclax induces Beclin 1- and ATG5-dependent apoptosis and autophagy in adenoid cystic carcinoma cells. Oral Dis. 2015, 21, 470–477. [Google Scholar] [CrossRef] [PubMed]
  144. McCoy, F.; Hurwitz, J.; McTavish, N.; Paul, I.; Barnes, C.; O’Hagan, B.; Odrzywol, K.; Murray, J.; Longley, D.; McKerr, G.; et al. Obatoclax induces Atg7-dependent autophagy independent of beclin-1 and BAX/BAK. Cell. Death Dis. 2010, 1, e108. [Google Scholar] [CrossRef] [PubMed]
  145. Tang, Y.; Hamed, H.A.; Cruickshanks, N.; Fisher, P.B.; Grant, S.; Dent, P. Obatoclax and lapatinib interact to induce toxic autophagy through NOXA. Mol. Pharmacol. 2012, 81, 527–540. [Google Scholar] [CrossRef] [PubMed]
  146. Wei, Y.; Kadia, T.; Tong, W.; Zhang, M.; Jia, Y.; Yang, H.; Hu, Y.; Viallet, J.; O’Brien, S.; Garcia-Manero, G. The combination of a histone deacetylase inhibitor with the BH3-mimetic GX15-070 has synergistic antileukemia activity by activating both apoptosis and autophagy. Autophagy 2010, 6, 976–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Stamelos, V.A.; Fisher, N.; Bamrah, H.; Voisey, C.; Price, J.C.; Farrell, W.E.; Redman, C.W.; Richardson, A. The BH3 Mimetic Obatoclax Accumulates in Lysosomes and Causes Their Alkalinization. PLoS ONE 2016, 11, e0150696. [Google Scholar] [CrossRef] [PubMed]
  148. Yu, L.; Wu, W.K.; Gu, C.; Zhong, D.; Zhao, X.; Kong, Y.; Lin, Q.; Chan, M.T.; Zhou, Z.; Liu, S. Obatoclax impairs lysosomal function to block autophagy in cisplatin-sensitive and -resistant esophageal cancer cells. Oncotarget 2016, 7, 14693–14707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Basit, F.; Cristofanon, S.; Fulda, S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 2013, 20, 1161–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Chen, S.; Wang, G.; Niu, X.; Zhao, J.; Tan, W.; Wang, H.; Zhao, L.; Ge, Y. Combination of AZD2281 (Olaparib) and GX15-070 (Obatoclax) results in synergistic antitumor activities in preclinical models of pancreatic cancer. Cancer Lett. 2014, 348, 20–28. [Google Scholar] [CrossRef] [PubMed]
  151. Zhao, X.Y.; Lin, Q.H.; Que, F.C.; Gu, C.P.; Yu, L.; Liu, S.W. Synergistic anti-tumor effect of obatoclax and MG-132 in esophageal cancer cell line CaES-17. Nan Fang Yi Ke Da Xue Xue Bao 2016, 36, 506–513. [Google Scholar] [PubMed]
  152. Li, J.; Viallet, J.; Haura, E.B. A small molecule pan-Bcl-2 family inhibitor, GX15-070, induces apoptosis and enhances cisplatin-induced apoptosis in non-small cell lung cancer cells. Cancer Chemother. Pharmacol. 2008, 61, 525–534. [Google Scholar] [CrossRef] [PubMed]
  153. Ma, J.; Zhao, Z.; Wu, K.; Xu, Z.; Liu, K. MCL-1 is the key target of adjuvant chemotherapy to reverse the cisplatin-resistance in NSCLC. Gene 2016, 587, 147–154. [Google Scholar] [CrossRef] [PubMed]
  154. Wei, W.J.; Sun, Z.K.; Shen, C.T.; Song, H.J.; Zhang, X.Y.; Qiu, Z.L.; Luo, Q.Y. Obatoclax and LY3009120 Efficiently Overcome Vemurafenib Resistance in Differentiated Thyroid Cancer. Theranostics 2017, 7, 987–1001. [Google Scholar] [CrossRef] [PubMed]
  155. Heidari, N.; Hicks, M.A.; Harada, H. GX15-070 (obatoclax) overcomes glucocorticoid resistance in acute lymphoblastic leukemia through induction of apoptosis and autophagy. Cell Death Dis. 2010, 1, e76. [Google Scholar] [CrossRef] [PubMed]
  156. Bonapace, L.; Bornhauser, B.C.; Schmitz, M.; Cario, G.; Ziegler, U.; Niggli, F.K.; Schafer, B.W.; Schrappe, M.; Stanulla, M.; Bourquin, J.P. Induction of autophagy-dependent necroptosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance. J. Clin. Investig. 2010, 120, 1310–1323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. O’Brien, S.M.; Claxton, D.F.; Crump, M.; Faderl, S.; Kipps, T.; Keating, M.J.; Viallet, J.; Cheson, B.D. Phase I study of obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 family antagonist, in patients with advanced chronic lymphocytic leukemia. Blood 2009, 113, 299–305. [Google Scholar] [CrossRef] [PubMed]
  158. Hrgovic, I.; Glavic, Z.; Kovacic, Z.; Mulic, S.; Zunic, L.; Hrgovic, Z. Repeated administration of inhibitors for ion pumps reduce markedly tumor growth in vivo. Med. Arch. 2014, 68, 76–78. [Google Scholar] [CrossRef] [PubMed]
  159. 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] [PubMed]
  160. Schimmer, A.D.; Raza, A.; Carter, T.H.; Claxton, D.; Erba, H.; DeAngelo, D.J.; Tallman, M.S.; Goard, C.; Borthakur, G. A multicenter phase I/II study of obatoclax mesylate administered as a 3- or 24-hour infusion in older patients with previously untreated acute myeloid leukemia. PLoS ONE 2014, 9, e108694. [Google Scholar] [CrossRef] [PubMed]
  161. Oki, Y.; Copeland, A.; Hagemeister, F.; Fayad, L.E.; Fanale, M.; Romaguera, J.; Younes, A. Experience with obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 family antagonist in patients with relapsed or refractory classical Hodgkin lymphoma. Blood 2012, 119, 2171–2172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Arellano, M.L.; Borthakur, G.; Berger, M.; Luer, J.; Raza, A. A phase II, multicenter, open-label study of obatoclax mesylate in patients with previously untreated myelodysplastic syndromes with anemia or thrombocytopenia. Clin. Lymphoma Myeloma Leuk 2014, 14, 534–539. [Google Scholar] [CrossRef] [PubMed]
  163. Langer, C.J.; Albert, I.; Ross, H.J.; Kovacs, P.; Blakely, L.J.; Pajkos, G.; Somfay, A.; Zatloukal, P.; Kazarnowicz, A.; Moezi, M.M.; et al. Randomized phase II study of carboplatin and etoposide with or without obatoclax mesylate in extensive-stage small cell lung cancer. Lung Cancer 2014, 85, 420–428. [Google Scholar] [CrossRef] [PubMed]
  164. Paik, P.K.; Rudin, C.M.; Pietanza, M.C.; Brown, A.; Rizvi, N.A.; Takebe, N.; Travis, W.; James, L.; Ginsberg, M.S.; Juergens, R.; et al. A phase II study of obatoclax mesylate, a Bcl-2 antagonist, plus topotecan in relapsed small cell lung cancer. Lung Cancer 2011, 74, 481–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Urtishak, K.A.; Edwards, A.Y.; Wang, L.S.; Hudome, A.; Robinson, B.W.; Barrett, J.S.; Cao, K.; Cory, L.; Moore, J.S.; Bantly, A.D.; et al. Potent obatoclax cytotoxicity and activation of triple death mode killing across infant acute lymphoblastic leukemia. Blood 2013, 121, 2689–2703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Boehmerle, W.; Endres, M. Salinomycin induces calpain and cytochrome c-mediated neuronal cell death. Cell Death Dis. 2011, 2, e168. [Google Scholar] [CrossRef] [PubMed]
  167. Delwar, Z.M.; Avramidis, D.; Siden, A.; Cruz, M.; Yakisich, J.S. Depletion of drug-surviving glioma cells by a second phase treatment with low concentration of salinomycin. Drugs Ther. Stud. 2011, 1, 21–25. [Google Scholar] [CrossRef]
  168. Huang, X.; Borgstrom, B.; Stegmayr, J.; Abassi, Y.; Kruszyk, M.; Leffler, H.; Persson, L.; Albinsson, S.; Massoumi, R.; Scheblykin, I.G.; et al. The Molecular Basis for Inhibition of Stemlike Cancer Cells by Salinomycin. ACS Cent. Sci. 2018, 4, 760–767. [Google Scholar] [CrossRef] [PubMed]
  169. Najumudeen, A.K.; Jaiswal, A.; Lectez, B.; Oetken-Lindholm, C.; Guzman, C.; Siljamaki, E.; Posada, I.M.; Lacey, E.; Aittokallio, T.; Abankwa, D. Cancer stem cell drugs target K-ras signaling in a stemness context. Oncogene 2016, 35, 5248–5262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. 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. Biol. Interact. 2015, 228, 100–107. [Google Scholar] [CrossRef] [PubMed]
  171. Jing, H.; Qu, X.; Liu, L.; Xia, H. A Novel Long Noncoding RNA (lncRNA), LL22NC03-N64E9.1, Promotes the Proliferation of Lung Cancer Cells and is a Potential Prognostic Molecular Biomarker for Lung Cancer. Med. Sci. Monit. 2018, 24, 4317–4323. [Google Scholar] [CrossRef] [PubMed]
  172. Chen, K.F.; Su, J.C.; Liu, C.Y.; Huang, J.W.; Chen, K.C.; Chen, W.L.; Tai, W.T.; Shiau, C.W. A novel obatoclax derivative, SC-2001, induces apoptosis in hepatocellular carcinoma cells through SHP-1-dependent STAT3 inactivation. Cancer Lett. 2012, 321, 27–35. [Google Scholar] [CrossRef] [PubMed]
  173. Su, J.C.; Chen, K.F.; Chen, W.L.; Liu, C.Y.; Huang, J.W.; Tai, W.T.; Chen, P.J.; Kim, I.; Shiau, C.W. Synthesis and biological activity of obatoclax derivatives as novel and potent SHP-1 agonists. Eur. J. Med. Chem. 2012, 56, 127–133. [Google Scholar] [CrossRef] [PubMed]
  174. Antoszczak, M.; Urbaniak, A.; Delgado, M.; Maj, E.; Borgstrom, B.; Wietrzyk, J.; Huczynski, A.; Yuan, Y.; Chambers, T.C.; Strand, D. Biological activity of doubly modified salinomycin analogs—Evaluation in vitro and ex vivo. Eur. J. Med. Chem. 2018, 156, 510–523. [Google Scholar] [CrossRef] [PubMed]
  175. Borgstrom, B.; Huang, X.; Chygorin, E.; Oredsson, S.; Strand, D. Salinomycin Hydroxamic Acids: Synthesis, Structure, and Biological Activity of Polyether Ionophore Hybrids. ACS Med. Chem. Lett. 2016, 7, 635–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Huczynski, A.; Janczak, J.; Antoszczak, M.; Wietrzyk, J.; Maj, E.; Brzezinski, B. Antiproliferative activity of salinomycin and its derivatives. Bioorg. Med. Chem. Lett. 2012, 22, 7146–7150. [Google Scholar] [CrossRef] [PubMed]
  177. Urbaniak, A.; Delgado, M.; Antoszczak, M.; Huczynski, A.; Chambers, T.C. Salinomycin derivatives exhibit activity against primary acute lymphoblastic leukemia (ALL) cells in vitro. Biomed. Pharmacother. 2018, 99, 384–390. [Google Scholar] [CrossRef] [PubMed]
  178. Zhang, W.; Wu, J.; Li, B.; Lian, X.; Xia, J.; Zhou, Q.; Wu, S. Design and synthesis of conformationally constrained salinomycin derivatives. Eur. J. Med. Chem. 2017, 138, 353–356. [Google Scholar] [CrossRef] [PubMed]
  179. Zhang, W.; Wu, J.; Li, B.; Wu, H.; Wang, L.; Hao, J.; Wu, S.; Zhou, Q. Structure-activity & structure-toxicity relationship study of salinomycin diastereoisomers and their benzoylated derivatives. Org. Biomol. Chem. 2016, 14, 2840–2845. [Google Scholar] [PubMed]
  180. Zhou, J.; Sun, J.; Chen, H.; Peng, Q. Promoted delivery of salinomycin sodium to lung cancer cells by dual targeting PLGA hybrid nanoparticles. Int. J. Oncol. 2018, 53, 1289–1300. [Google Scholar] [CrossRef] [PubMed]
  181. Chen, F.; Zeng, Y.; Qi, X.; Chen, Y.; Ge, Z.; Jiang, Z.; Zhang, X.; Dong, Y.; Chen, H.; Yu, Z. Targeted salinomycin delivery with EGFR and CD133 aptamers based dual-ligand lipid-polymer nanoparticles to both osteosarcoma cells and cancer stem cells. Nanomedicine 2018, 14, 2115–2127. [Google Scholar] [CrossRef] [PubMed]
  182. Mi, Y.; Huang, Y.; Deng, J. The enhanced delivery of salinomycin to CD133(+) ovarian cancer stem cells through CD133 antibody conjugation with poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles. Oncol. Lett. 2018, 15, 6611–6621. [Google Scholar] [CrossRef] [PubMed]
  183. Daman, Z.; Faghihi, H.; Montazeri, H. Salinomycin nanoparticles interfere with tumor cell growth and the tumor microenvironment in an orthotopic model of pancreatic cancer. Drug Dev. Ind. Pharm. 2018, 44, 1434–1442. [Google Scholar] [CrossRef] [PubMed]
  184. Choi, Y.J.; Gurunathan, S.; Kim, J.H. Graphene Oxide-Silver Nanocomposite Enhances Cytotoxic and Apoptotic Potential of Salinomycin in Human Ovarian Cancer Stem Cells (OvCSCs): A Novel Approach for Cancer Therapy. Int. J. Mol. Sci. 2018, 19, 710. [Google Scholar] [CrossRef] [PubMed]
  185. Yu, Z.; Chen, F.; Qi, X.; Dong, Y.; Zhang, Y.; Ge, Z.; Cai, G.; Zhang, X. Epidermal growth factor receptor aptamer-conjugated polymer-lipid hybrid nanoparticles enhance salinomycin delivery to osteosarcoma and cancer stem cells. Exp. Ther. Med. 2018, 15, 1247–1256. [Google Scholar] [CrossRef] [PubMed]
  186. Zeng, Y.B.; Yu, Z.C.; He, Y.N.; Zhang, T.; Du, L.B.; Dong, Y.M.; Chen, H.W.; Zhang, Y.Y.; Wang, W.Q. Salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamers selectively suppress human CD20+ melanoma stem cells. Acta Pharmacol. Sin. 2018, 39, 261–274. [Google Scholar] [CrossRef] [PubMed]
  187. Zhang, Y.; Zhang, Q.; Sun, J.; Liu, H.; Li, Q. The combination therapy of salinomycin and gefitinib using poly(d,l-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles for targeting both lung cancer stem cells and cancer cells. Onco Targets Ther. 2017, 10, 5653–5666. [Google Scholar] [CrossRef] [PubMed]
  188. Li, J.; Xu, W.; Yuan, X.; Chen, H.; Song, H.; Wang, B.; Han, J. Polymer-lipid hybrid anti-HER2 nanoparticles for targeted salinomycin delivery to HER2-positive breast cancer stem cells and cancer cells. Int. J. Nanomed. 2017, 12, 6909–6921. [Google Scholar] [CrossRef] [PubMed]
  189. Li, L.; Cui, D.; Ye, L.; Li, Y.; Zhu, L.; Yang, L.; Bai, B.; Nie, Z.; Gao, J.; Cao, Y. Codelivery of salinomycin and docetaxel using poly(d,l-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles to target both gastric cancer cells and cancer stem cells. Anticancer Drugs 2017, 28, 989–1001. [Google Scholar] [CrossRef] [PubMed]
  190. Gong, Z.; Chen, D.; Xie, F.; Liu, J.; Zhang, H.; Zou, H.; Yu, Y.; Chen, Y.; Sun, Z.; Wang, X.; et al. Codelivery of salinomycin and doxorubicin using nanoliposomes for targeting both liver cancer cells and cancer stem cells. Nanomedicine (Lond.) 2016, 11, 2565–2579. [Google Scholar] [CrossRef] [PubMed]
  191. Zhao, P.; Xia, G.; Dong, S.; Jiang, Z.X.; Chen, M. An iTEP-salinomycin nanoparticle that specifically and effectively inhibits metastases of 4T1 orthotopic breast tumors. Biomaterials 2016, 93, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. 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] [PubMed]
  193. Tigli Aydin, R.S.; Kaynak, G.; Gumusderelioglu, M. Salinomycin encapsulated nanoparticles as a targeting vehicle for glioblastoma cells. J. Biomed. Mater. Res. A 2016, 104, 455–464. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Ionophore mediated ion transport across the membrane. (A) Small ionophores “ion carriers” bind with ion, shield it from lipophilic interior of membrane, transport it across the membrane and release it other side of membrane. (B) Large ionophores form “ion channels” across the membrane and transport ions through these channels. These channels have a hydrophilic interior which assist in transport of ions while its lipophilic exterior shield ions from repulsive interior of membrane. (C) Polyether ionophores carry ions across membrane by electroneutral, electrogenic and biomimetic methods based on the microenvironmental conditions and structure of ionophore. Panel C was modified from [71].
Figure 1. Ionophore mediated ion transport across the membrane. (A) Small ionophores “ion carriers” bind with ion, shield it from lipophilic interior of membrane, transport it across the membrane and release it other side of membrane. (B) Large ionophores form “ion channels” across the membrane and transport ions through these channels. These channels have a hydrophilic interior which assist in transport of ions while its lipophilic exterior shield ions from repulsive interior of membrane. (C) Polyether ionophores carry ions across membrane by electroneutral, electrogenic and biomimetic methods based on the microenvironmental conditions and structure of ionophore. Panel C was modified from [71].
Cancers 10 00360 g001
Table 1. Ionophores with anticancer activity.
Table 1. Ionophores with anticancer activity.
IonophoreTransported IonCancer TypeTarget CSCsReference
NigericinK+, H+SeveralYes[4,5,6]
SalinomycinK+, Ca2+SeveralYes[7,8,9]
ObatoclaxHCO3, ClSeveralYes[10,11,12,13,14,15]
GramicidinH+, Na+, K+Renal cell carcinoma,Not known[16,17]
IonomycinCa2+BreastNot known[18,19]
MonensinNa+, H+Glioblastoma, BladderNot known[20,21,22]
ValinomycinK+Ovarian, Colorectal,Likely[3,23,24]
LasalocidK+, Na+, Ca2+,ProstateNot known[25]
EnniatinMg2+Colon, OvarianNot known[26,27]
BeauvericinNH4+, Ca2+, Ba2+Prostate
Cervical, Colorectal Hepatoma, Lung
Not known[28,29]
Table 2. Registered clinical trials (www.clinicaltrials.gov) and published results.
Table 2. Registered clinical trials (www.clinicaltrials.gov) and published results.
IonophoreConditionOther DrugsPhaseClinical TrialPublished Results
Salinomycin-
Nigericin-
ObatoclaxAML- NCT00684918[160]
Chronic Lymphocytic Leukemia-I/IINCT00600964[157]
Extensive-stage Small-Cell Lung CancerCarboplatin/etoposideI/IINCT00682981[163]
Lymphoma, FollicularRituximab NCT00427856-
Non-Hodgkin Lymphoma
Recurrent Adult Diffuse Large Cell Lymphoma
Recurrent Grade 1 Follicular Lymphoma
(and 5 more…)
Bortezomib NCT00538187-
B-cell Chronic Lymphocytic
Leukemia
Leukemia
Prolymphocytic Leukemia
(and 5 more…)
Fludarabine rituximab NCT00612612-
Leukemia
Systemic Mastocytosis
NCT00918931-
Mantle-Cell LymphomaBortezomib NCT00407303-
Hodgkin’s Lymphoma NCT00359892[161]
Extensive-stage Small-Cell Lung CancerCarboplatine and Etoposide NCT01563601-
Lung CancerDocetaxel NCT00405951-
Myelodysplastic Syndromes NCT00413114[162]
Acute Leukemias of Ambiguous Lineage
Acute Undifferentiated Leukemia
Angioimmunoblastic T-cell Lymphoma
(and 26 more…)
Dexrazoxane hydrochloride
Doxorubicin hydrochloride
(and 3 more…)
NCT00933985[165]
Refractory Multiple Myeloma
Stage I Multiple Myeloma
Stage II Multiple Myeloma
Stage III Multiple Myeloma
Bortezomib NCT00719901-
Recurrent Small-Cell Lung Cancer
Unspecified Adult Solid Tumor
Topotecan hydrochloride NCT00521144[164]
Extranodal Marginal Zone B-cell Lymphoma of Mucosa-associated Lymphoid Tissue
Nodal Marginal Zone B-cell Lymphoma
Recurrent Grade 1 Follicular Lymphoma
(and 4 more…)
Bendamustine hydrochloride NCT01238146-
Myelofibrosis NCT00360035-
Hematological Malignancies NCT00438178-
Leukemia (samples) NCT01150656-
Metastatic MelanomaTemozolomideI/IINCT00724841-
Table 3. Delivery of Salinomycin using nanoparticles.
Table 3. Delivery of Salinomycin using nanoparticles.
NanoparticleCancer TypeEfficacyReference
SS lipid-polymer hybrid nanoparticlesLung[180]
CESP *Osteosarcoma[181]
CD133-SAL-NPCD133+ ovarian cancer stem cells and nude mice bearing ovarian cancer xenografts[182]
Poly (lactic-co-glycolic acid) (PLGA) nanoparticlesPancreatic cancerBlocked tumor growth by 52% compared to the control.[183]
rGO-AgHuman ovarian cancer stem cells[184]
EGFR-SNPsOsteosarcoma and cancer stem cells[185]
CD20-SA-NPsHuman CD20+ melanoma stem cells[186]
Salinomycin-NPs + gefitinib-NPsLung cancer and lung cancer stem cells[187]
Sali-NP-HER2HER2-positive breast cancer stem cells and cancer cells[188]
Salinomycin-NPs + docetaxel-NPsGastric cancer cells and cancer stem cells[189]
SDLNLiver cancer cells and cancer stem cells[190]
iTEP-Sali-ABA NP + iTEP NP-delivered paclitaxelMetastases of 4T1 orthotopic breast tumors[191]
Salinomycin-NPs + Paclitaxell-NPsBreast cancer stem cells and cancer cells[192]
P80-SAL-PLGAGlioblastoma[193]
CESNHepatocellular carcinoma[115]
Ap-SAL-NPOsteosarcoma cancer stem cells[118]
* CESP = salinomycin-entrapped lipid-polymer nanoparticles labeled with CD133 and EGFR aptamers; CD133-SAL-NP = salinomycin-loaded poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles conjugated with CD133 antibodies; rGO-Ag = reduced graphene oxide-silver nanoparticle nanocomposites; EGFR-SNPs = EGFR aptamer-conjugated salinomycin-loaded polymer-lipid hybrid nanoparticles; CD20-SA-NPs = salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamer; Sali-NP-HER2 = salinomycin-loaded polymer-lipid hybrid anti-HER2 nanoparticles; SDLN = salinomycin and doxorubicin nanoliposomes; iTEP-Sali-ABA NP = immune-tolerant, elastin-like polypeptide (iTEP)-based nanoparticle; P80-SAL-PLGA = Salinomycin-encapsulated polysorbate 80-coated poly(lactic-co-glycolic acid) nanoparticles; CESN = salinomycin-loaded poly(lactic-co-glycolic acid) nanoparticles conjugated with both CD133 aptamers A15 and EGFR aptamers CL4; Ap-SAL-NP = salinomycin-loaded PEGylated poly(lactic-co-glycolic acid) nanoparticles (SAL-NP) conjugated with CD133 aptamers.

Share and Cite

MDPI and ACS Style

Kaushik, V.; Yakisich, J.S.; Kumar, A.; Azad, N.; Iyer, A.K.V. Ionophores: Potential Use as Anticancer Drugs and Chemosensitizers. Cancers 2018, 10, 360. https://doi.org/10.3390/cancers10100360

AMA Style

Kaushik V, Yakisich JS, Kumar A, Azad N, Iyer AKV. Ionophores: Potential Use as Anticancer Drugs and Chemosensitizers. Cancers. 2018; 10(10):360. https://doi.org/10.3390/cancers10100360

Chicago/Turabian Style

Kaushik, Vivek, Juan Sebastian Yakisich, Anil Kumar, Neelam Azad, and Anand K. V. Iyer. 2018. "Ionophores: Potential Use as Anticancer Drugs and Chemosensitizers" Cancers 10, no. 10: 360. https://doi.org/10.3390/cancers10100360

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop