Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

Benzoxazole-based Zn(II) and Cu(II) Complexes Overcome Multidrug-resistance in Cancer

GABRIELLA SPENGLER, ANNAMÁRIA KINCSES, BÁLINT RÁCZ, BORISZ VARGA, GENKI WATANABE, RYOSUKE SAIJO, HIROSHI SEKIYA, EIJI TAMAI, JUN MAKI, JOSEPH MOLNÁR and MASAMI KAWASE
Anticancer Research November 2018, 38 (11) 6181-6187; DOI: https://doi.org/10.21873/anticanres.12971
GABRIELLA SPENGLER
1Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ANNAMÁRIA KINCSES
1Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
BÁLINT RÁCZ
1Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
BORISZ VARGA
1Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
2Department of Rheumatology and Immunology, University of Szeged, Szeged, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
GENKI WATANABE
3Faculty of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
RYOSUKE SAIJO
3Faculty of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROSHI SEKIYA
4Department of Infectious Diseases, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
EIJI TAMAI
4Department of Infectious Diseases, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JUN MAKI
4Department of Infectious Diseases, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JOSEPH MOLNÁR
1Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MASAMI KAWASE
3Faculty of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: kawase@g.matsuyama-u.ac.jp
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Multidrug resistance (MDR) represents a significant impediment to successful cancer treatment. In this study, novel metal [Zn(II), Cu(II), Mg(II), Ni(II), Pd(II), and Ag(I)] complexes of 2-trifluoroacetonylbenzoxazole previously synthesized and characterized by our group were tested for their MDR-reversing activity in comparison with the free ligands in L5178Y mouse T-lymphoma (MDR) cells transfected with human ATP-binding cassette sub-family B member 1 (ABCB1; P-glycoprotein) gene. Materials and Methods: Cytotoxic and antiproliferative effects of the complexes were assessed by the thiazolyl blue tetrazolium bromide (MTT) method. Modulation of ABCB1 activity was measured by rhodamine 123 accumulation assay using flow cytometry. The apoptosis-inducing activity of some complexes was also tested on the multidrug resistant L5178Y mouse T-lymphoma cells, using the annexin-V/propidium iodide assay. Results: When compared to the free ligand, a remarkable enhancement in MDR reversal and cytotoxic activity was found for the Zn(II) and Cu(II) complexes. The activity of the complexes proved to be up to 29- and 5-fold higher than that of the ligands and the ABCB1 inhibitor verapamil as positive control, respectively. The complexes possessed a remarkable potential to induce apoptosis of MDR cells. Conclusion: Our results suggest that the Zn(II) and Cu(II) complexes display significant MDR-reversing activity in a dose-dependent manner and possess strong cytotoxic activity and a remarkable potential to induce apoptosis in MDR L5178Y mouse T-lymphoma cells.

  • Multidrug resistance
  • metal complex
  • benzoxazole
  • ABCB1
  • P-glycoprotein
  • mouse T-lymphoma
  • apoptosis

Multidrug resistance (MDR) of cancer is defined as the insensitivity of cancer cells to the cytotoxic action of various anticancer drugs (1-4). One of the major mechanisms of MDR is the overexpression of efflux pumps, which reduces the accumulation of administered anticancer drugs. Multidrug efflux pumps fall into one of six distinct families of membrane proteins, with the most important MDR transporters of cancer cells belonging to the ATP-binding cassette (ABC) transporters, such as P-glycoprotein (P-gp; ABCB1), multidrug resistance-associated protein-1 (ABCC1) and breast cancer resistance protein (ABCG2) (5).

One promising approach to overcome MDR mediated by P-gp is based on the development of MDR-reversal agents that possess efflux pump inhibitor properties (6-8). A number of MDR-reversal agents of synthetic or natural origin have been reported and some of them have reached the stage of clinical trials. However, none of them has been approved for clinical use because of undesirable side-effects (9, 10). Therefore, it is important to continue research in this area in order to discover new inhibitors of primary efflux pumps.

The discovery and development of new metal-based chemotherapeutics is an ever-growing area of research in medicinal chemistry. Coordination of organic compounds with metal causes drastic changes in the biological properties of both the ligand and the metal moiety. Therefore, many metal complexes had been designed and synthesized in order to explore their pharmacological and antitumor activity (11-14). The platinum-based drug cisplatin is one of the commonly used drugs in the clinic in the treatment of various types of human cancer, but its serious side-effects and resistance by cancer cells confine its clinical application (15). In order to curb increasing resistance, it is a necessityto design potential alternatives. Recently, several ruthenium-based complexes, such as NAMI-A, KP1019, KP1339, and TD1433, have been developed, of which the latter two are currently undergoing clinical trials (16, 17). In this endeavor, transition metals such as Cu, Zn, Ni, Ag, and Au are considered to be promising candidates for designing such therapeutic agents because of their biocompatibility and their endogenous presence in living system as co-factors in several enzymes (18-21).

The benzoxazole heterocycle system is an important pharmacophore and privileged structure in medicinal chemistry (22, 23). Its derivatives, and particularly, 2-substituted benzoxazoles, exert various biological activities such as anticancer, antimicrobial, antiviral, and antifungal (24-28).

In previous studies, we synthesized new Zn(II), Mg(II), Ni(II), Cu(II), Pd(II), and Ag(I) complexes of 2-trifluoroacetonylbenzoxazole ligand, of which the later displayed significant antibacterial activities (29). In the present study, 2-trifluoroacetonylbenzoxazoles were used as active ligands to coordinate with transition metals because these compounds are highly desirable ligands for their ability to coordinate with metal center, good planarity, highly hydrophobic DNA interactions and mimicry of the purine nucleus (30, 31).

In order to identify specific and selective MDR reversal agents, we have evaluated the cytotoxic and MDR reversing potential of three ligands (compounds 1-3, Figure 1) and their eight metal complexes (compounds 4-11; Figure 1) in parental L5178Y mouse T-lymphoma cell line and its human ABCB1 gene-transfected (MDR) subline. In addition, the capacity of potent MDR modulators as apoptosis inducers was also evaluated.

In this article, we report the MDR-modulating activities of metal complexes and the identification of selective MDR modulators of the ABCB1 pump in cancer cells.

Materials and Methods

Compounds. The syntheses of the ligands and metal complexes were described previously (29) and their structures are shown in Figure 1. The compounds were dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich, Madrid, Spain).

Cell lines. The L5178Y mouse T-cell lymphoma cells (FDA, Silver Spring, MD, USA) were transfected with pHaMDR1/A retrovirus (7). The ABCB1-expressing MDR cell line was selected by culturing the infected cells with colchicine. Parental L5178Y mouse T-cell lymphoma cells and the human ABCB1-transfected subline were cultured in McCoy's 5A medium supplemented with 10% heat inactivated horse serum, 200 mM L-glutamine, and penicillin–streptomycin mixture at 100 U/l and 10 mg/l, respectively.

Assay for cytotoxic effect. The effects of increasing concentrations of the compounds on cell growth were tested in 96-well flat-bottomed microtiter plates. The compounds were diluted in 100 μI of McCoy's 5A medium then 1×104 cells in 100 μI of medium were added to each well, with the exception of the medium control wells. The culture plates were further incubated at 37°C for 24 h. At the end of the incubation period, 20 μI of thiazolyl blue tetrazolium bromide (MTT; Sigma-Aldrich) solution (from a 5 mg/ml stock) were added to each well. After incubation at 37°C for 4 h, 100 μI of sodium dodecyl sulfate (Sigma-Aldrich) solution (10% in 0.01 M HCl) were added to each well and the plates were further incubated at 37°C overnight. Cell growth was determined by measuring the optical density (OD) at 540 nm (ref. 630 nm) with a Multiscan EX ELISA reader (Thermo Labsystems, Cheshire, WA, USA) (7). Inhibition of cell growth was determined according to the following formula: Embedded Image Eq 1 Where IC50 was defined as the inhibitory dose that reduce the growth of the cells exposed to the compound by 50%.

Fluorescence uptake assay. The density of the L5178Y parental and MDR cell lines were adjusted to 2×106 cells/mI, re-suspended in serum-free McCoy's 5A medium and distributed in 0.5 mI aliquots into Eppendorf tubes. The test compounds were added at a final concentration of 2 and 20 μM and the samples were incubated for 10 min at room temperature. Verapamil (Sigma-Aldrich) was applied as positive control at 20 μM. DMSO was added to the negative control tubes in the same volume as had been used for the tested compounds. No activity of DMSO was observed. Next, 10 μl (5.2 μM final concentration) of rhodamine 123 (Sigma-Aldrich) were added to the samples and the cells were incubated for a further 20 min at 37°C, washed twice and re-suspended in 1 mI phosphate buffered saline for analysis. The fluorescence of the cell population was measured with a PartecCyFlow® flow cytometer (Partec, Münster, Germany). The percentage of mean fluorescence intensity was calculated for the treated MDR cells as compared with untreated cells (7). A fluorescence activity ratio (FAR) was calculated on the basis of the measured fluorescence values via the following equation: Embedded Image Eq 2

Apoptosis assay. The assay was carried out using Annexin V-fluorescein isothiocyanate (FITC) Apoptosis Detection Kit (Calbiochem, Merck KGaA, Darmstadt, Germany) according to the manufacturer's instructions. The density of the mouse MDR T-lymphoma cell suspension was adjusted to approximately 1×106 cells/ml. The cell suspension was distributed into 0.5 ml aliquots (5×105 cells) to a 24-well microplate and the compounds were added at a final concentration of 10 μM. The apoptosis inducer 12H-benzo[a]phenothiazine (M627) was applied as positive control at a final concentration of 20 μM (32). The cells were incubated in the presence of the compounds for 1 h in a water-bath (37°C). The samples were then centrifuged and the culture medium was removed, the cells were washed with phosphate-buffered saline and fresh medium was added to the cells. The 24-well plates were incubated overnight at 37°C with 5% CO2. On the following day, the samples were then centrifuged at 2000 × g for 2 min at room temperature and the supernatant was removed and the cells were re-suspended in 0.5 mI fresh serum-free medium. After this procedure, the apoptosis assay was carried out according to the rapid protocol of the kit by which cells were stained with FITC-labelled annexin V and propidium iodide (PI) and then analyzed by flow cytometry. This discriminated between necrotic (annexin V−/PI+), viable (annexin V−/PI−), early apoptotic (annexin V+/PI−), and late apoptotic (annexin V+/PI+) cells. The fluorescence of cell populations was analyzed immediately using a PartecCyFlow® flow cytometer.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

The structures and molecular weights of the ligands (compounds 1-3) and metal complexes (compounds 4-11).

Results

We screened our in-house library of metal complexes and related compounds, most of them previously synthesized in our laboratory (29). The metal complexes 4-11 were easily prepared in moderate to good yields by the reaction of 2-trifluoroacetonylbenzoxazole ligands (compounds 1-3) with metal salts (29). The structures of compounds 1-11 screened for their MDR-modulating activities are shown in Figure 1

The cytotoxic activity of ligands and their metal complexes was evaluated using MTT assay in two tumor cell lines, parental L5178Y mouse T-lymphoma cells and its human ABCB1-gene transfected MDR subline. These MDR cells overexpress the ABCB1 protein that is responsible for drug efflux. The IC50 values are presented in Table I, in which the chemotherapeutic drug cisplatin was used as positive control. The results indicated that the cytotoxicity of Zn(II) complexes 4-6 was greater than those of the ligands 1-3. This observation suggested the advantages of Zn(II) complexes over their parent ligands in practical applications as MDR-modulating agents. The selectivity index (SI) for each compound was calculated based upon the comparison of the sensitivity (IC50) of parental (chemosensitive) to MDR (chemoresistant due to overexpression of ABCB1) cell lines. When the SI value is greater than 1, resistance in the MDR cells is reversed by a compound. Thus, selective cytotoxic activity was observed in the case of compounds 4 (SI=6.9), 5 (SI=2.7), 3 (SI=2.6), 2 (SI=2.3) and 9 (SI=2.1) in the MDR cells relative to the parental cells, suggesting that the compounds were able to modulate or block the ABCB1 transporter. On the other hand, compounds 1, 6, 7, 8, and 11 were equally active against both parental and MDR because the SI did not show any significant discrimination between parental and MDR cell lines. The Pd(II) complex 10 was not toxic against the MDR cells (SI<0.20). All compounds except Mg(II) complex 9 and Pd(II) complex 10 were more cytotoxic against both parental and MDR cells than cisplatin used as a positive control. In particular, the cytotoxicity of the Cu(II) complex 7 and Ag(I) complex 11 against the parental and the MDR cells was up to 7-to 9-fold higher than that of cisplatin.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Cytotoxic activity of ligands 1-3 and metal complexes 4-11 on parental and multidrug-resistant (MDR) cancer cells.

The rhodamine 123 (a substrate of ABCB1) exclusion assay was used to assess the potential ABCB1-mediated MDR-reversing activity of compounds 1-11. In this assay, the FAR was evaluated for the accumulation ratio of rhodamine 123 in MDR and parental cells (Table II). The reversal of MDR means that the accumulation of rhodamine 123 by the MDR cells should be similar to the accumulation by the chemosensitive parental cells, and the FAR should therefore be greater than 1. Verapamil, a well-known modulator (33), was used as positive control. The data in Table II reveal that complexes of Zn(II) (compounds 4, 5 and 6), Cu(II) (compound 7) and Ag(I) (compound 11) were potent MDR-reversing agents, while the ligands (compounds 1, 2 and 3) and complexes of Ni(II), Mg(II) and Pd(II) (compounds 8, 9 and 10) were inactive. It is noted that compounds 4 (FAR=28.0), 6 (FAR=16.9) and 7 (FAR=19.3) displayed a remarkable inhibition of MDR at a concentration of 20 μM. To study whether MDR reversal activity had a dose–effect dependence, four potent compounds 4-7 were tested at several concentrations (2, 10, 15 and 20 μM) and their FARs are presented in Table III. At 2 μM, these compounds were weak modulators. However, at 10-20 μM, the FAR values of these compounds were higher than 10 (11.0-29.4), leading us to classify them as strong MDR modulators (34). These results are of particular interest, since the MDR-reversing activity of verapamil at 20 μM is approximately 5. In addition, for compounds 5, 6 and 7, the effect was somewhat lower at higher concentration, meaning that the applied concentrations exceeded an optimum substrate concentration. The maximum effect was found at the lowest dose applied.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Multidrug resistance (MDR)-reversal activities of ligands 1-3 and metal complexes 4-11 on MDR cancer cells. The inhibition of ABCB1 transporter is evident when the fluorescence activity ratio (FAR) exceeds 1.

Next, the effect of some metal complexes (compounds 4-7 and 9-11) on apoptosis induction in the MDR cells was studied. An apoptosis inducer, M627, was used as the positive control (32). The compounds were tested at 10 μM and the results are summarized in Table IV. Complexes 5, 6 and 7 had the capacity to induce early apoptosis. Several complexes induced early apoptosis with potency decreasing in the order 11>M627>6>7>5. Late apoptosis was induced by compounds 4, 5, 6, 7, and 11: the most potent compounds were 5 and 6 as they induced late apoptosis in 69.6% and 59.1% of the cell population, respectively. Complexes 4, 5, 6, 7, and 11 led to cell death of 5.58%, 4.12%, 4.53%, 2.81%, and 3.81%, respectively. The Ag(I) complex 11 was able to induce significant apoptosis, similar to the potent MDR modifier M627. Apoptosis induction by Mg(II) complex 9 and Pd(II) complex 10 was less pronounced compared to the effective apoptosis inducers 4, 5, 6, and 7. These data show that the potent MDR modulators possessed a remarkable potential to induce apoptosis in MDR cells.

Discussion

The problem in cancer treatment is the frequent occurrence of MDR. A recent approach in therapy is to develop tailored treatment protocols targeting specific pathways of cellular proliferation or MDR. Therefore, an urgent task is to find more effective, modulators of low toxicity to inhibit MDR. A number of metal complexes, for examples, ruthenium complexes (35) and Cu(II), Zn(II), and Mn(II) N-(2-hydroxyacetophenone)glycinates (11) have also been shown to be strong MDR-reversing agents. In spite of advances in development of a number of good candidates for modulating MDR, we are still far away from being able to conclude that these agents would be applied clinically.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table III.

Fluorescence activity ratio (FAR) values for metal complexes 4-7 on multidrug-resistant (MDR) cancer cells at different concentrations. The inhibition of ABCB1 transporter is evident when FAR exceeds 1.

Previously, we reported that the ligand 1 had showed no obvious cytotoxicity [50% cytotoxicity concentration (CC50)=630 μM] against human gingival fibroblasts and was more cytotoxic to two human oral tumor cell lines (HSC-2: CC50=43 μM vs. 4.1 μM for doxorubicin; and HSG: CC50=79 μM) (36). In the present investigation, we developed new metal coordination compounds of the ligand 1 with the aim of obtaining suitable MDR-reversing agents. As a result of screening to obtain possible lead structures bearing metal, the strongest ability as MDR reversers and a many-fold activity when compared to verapamil was found for Zn(II) and Cu(II) complexes. Thus, the three parent ligands 1-3 showed no obvious MDR-modulating activity, while complexes 4-6 displayed a remarkable inhibition of MDR, indicating that ligand coordination plays an important role in the active complexes. Such increased activity of the metal complexes can be explained on the basis of the Overtone concept and Tweedy chelation theory (37-41). It is important to note that ABC transporters are primary efflux pumps deriving their energy from the hydrolysis of ATP. MDR modulators are believed to bind to the transmembrane domains of ABCB1 which leads to inhibition of ABC transporters (42-44). A number of studies revealed that lipophilicity is an important factor which controls MDR-modulating activity (6, 45). On chelation, the polarity of the metal ion is reduced to a greater extent due to the overlapping of ligand orbital and partial sharing of the positive charge of the metal ion with donor groups. Moreover, delocalization of the π electrons over the whole chelate ring is increased and lipophilicity enhances penetration of the complexes into the transmembrane domains of ABCB1. Therefore, a significant increase in MDR-modulating activity with Zn(II) and Cu(II) complexes can be explained on the basis of the above described Overtone concept and Tweedy chelation theory.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table IV.

Effect of compounds 4-7 and 9-11 (at 10 μM) on apoptosis induction in multidrug-resistant (MDR) cancer cellsa.

Conclusion

This study has identified a novel class of ABCB1 inhibitors, Zn(II) and Cu(II) complexes. These compounds demonstrate high potency against ABCB1 which far exceed that of the reference drug verapamil. We can conclude that the Zn(II) and Cu(II) complexes can not only be considered as effective anti-MDR agents, but also as apoptosis inducers and may be attractive lead compounds for further development as MDR-reversing agents. Hence, these potent metal complexes should be pursued in a variety of directions. Firstly, molecular mechanisms by which metal complexes bind to ABCB1 should be investigated, which will give insight for further designing of potent ABCB1 inhibitors. Among various metal ions, copper and zinc play a prominent role in the active sites of many metalloproteins. Studies have indicated that many zinc and copper complexes exhibit favorable antiproliferative activity against tumor cells, which have been attributed to their interaction with DNA (although their ability to bind to DNA largely depends on the coordinated ligand) (19-21). Secondly, evaluation of the activity of metal complexes against other efflux transporters such as MRP and BCRP should be carried out. Thirdly, the substituents of the benzoxazole ring in the ligand should be evaluated for MDR-modulating properties and selective toxicity for neoplasms.

The ease of synthesis and the small size of these compounds make them potential candidates for structure–activity relationships as efflux pump inhibitors in multidrug efflux systems.

Acknowledgements

This research was supported by the Szeged Foundation for Cancer Research, the European Union and the State of Hungary, co-financed by the European Social Fund in the framework of TÁMOP 4.2.4. A/2-11-1-2012-0001 ‘National Excellence Program’. Gabriella Spengler was supported by the János Bolyai Research Scholarship of the Hungarian Academy of Sciences. This study was supported by the project GINOP-2.3.2-15-2016-00038.

  • Received October 2, 2018.
  • Revision received October 15, 2018.
  • Accepted October 16, 2018.
  • Copyright© 2018, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

References

  1. ↵
    1. Robert J,
    2. Jarry C
    : Multidrug resistance-reversal agents. J Med Chem 46: 4805-4817, 2003.
    OpenUrlCrossRefPubMed
    1. Colabufo NA,
    2. Berardi F,
    3. Cantore M,
    4. Contino M,
    5. Inglese C,
    6. Niso M,
    7. Perrone R
    : Perspectives of P-glycoprotein-modulating agents in oncology and neurodegenerative diseases: pharmaceutical, biological, and diagnostic potentials. J Med Chem 53: 1883-1897, 2010.
    OpenUrlPubMed
    1. Zhang L,
    2. Ma S
    : Efflux pump inhibitors: a strategy to combat P-glycoprotein and the NorA multidrug-resistance pump. Chem Med Chem 5: 811-822, 2010.
    OpenUrlPubMed
  2. ↵
    1. Shukla S,
    2. Ohnuma S,
    3. Ambudkar SV,
    4. Nikaido H,
    5. Pages JMB
    : Improving cancer chemotherapy with modulators of ABC drug transporters. Curr Drug Targets 12: 621-630, 2011.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Gu X,
    2. Ren Z,
    3. Peng H,
    4. Peng S,
    5. Zhang Y
    : Bifendate-chalcone hybrids: A new class of potential dual inhibitors of P-gylcoprotein and breast cancer resistance protein. Biochem Biophys Res Commun 455: 318-322, 2014.
    OpenUrl
  4. ↵
    1. Spengler G,
    2. Ocsovszki I,
    3. Tonki AS,
    4. Saijo R,
    5. Watanabe G,
    6. Kawase M,
    7. Molnar J
    : Fluorinated beta-di-keto phosphorus ylides are novel inhibitors of the ABCB1 efflux pump of cancer cells. Anticancer Res 35: 5915-5920, 2015.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Paterna A,
    2. Kincses A,
    3. Spengler G,
    4. Mulhovo S,
    5. Molnar J,
    6. Ferreira MU
    : Dregamine and tabernaemontanine derivatives as ABCB1 modulators on resistant cancer cells. Eur J Med Chem 128: 247-257, 2017.
    OpenUrl
  6. ↵
    1. Bourichi S,
    2. Misbahi H,
    3. Rodi YK,
    4. Chahdi FO,
    5. Essassi EM,
    6. Szabó S,
    7. Szalontai B,
    8. Gajdács M,
    9. Molnár J,
    10. Spengler G
    : In vitro evaluation of the multidrug resistance reversing activity of novel imidazo[4,5-b]pyridine derivatives. Anticancer Res 38: 3999-4003, 2018.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Klinkhammer W,
    2. Muller H,
    3. Globisch C,
    4. Pajeva IK,
    5. Wiese M
    : Synthesis and biological evaluation of a small molecule library of 3rd-generation multidrug-resistance modulators. Bioorg Med Chem 17: 2524-2535, 2009.
    OpenUrlPubMed
  8. ↵
    1. Singh S,
    2. Prasad NR,
    3. Chufan EE,
    4. Patel BA,
    5. Wang Y-J,
    6. Chen Z-S,
    7. Ambudkar SV,
    8. Talele TT
    : Design and synthesis of human ABCB1 (P-glycoprotein) inhibitors by peptide coupling of diverse chemical scaffolds on carboxyl and amino termini of (S)-valine-derived thiazole amino acid. J Med Chem 57: 4058-4072, 2014.
    OpenUrlPubMed
  9. ↵
    1. Ganguly A,
    2. Charaborty P,
    3. Banerjee K,
    4. Choudhuri SK
    : The role of a Schiff base scaffold, N-(2-hydroxy acetophenone) glycinate-in overcoming multidrug resistance in cancer. Eur J Pharm Sci 51: 96-109, 2014.
    OpenUrl
    1. Turel I
    : Special issue: Practical applications of metal complexes. Molecules 20: 7951-7956, 2015.
    OpenUrl
    1. Allardyce CS,
    2. Dyson PJ
    : Metal-based drugs that break the rules. Dalton Trans 45: 3201-3209, 2016.
    OpenUrl
  10. ↵
    1. Zhang P,
    2. Sadler PJ
    : Advances in the design of organometallic anticancer complexes. J Organometal Chem 839: 5-14, 2017.
    OpenUrl
  11. ↵
    1. Lazarevic T,
    2. Rilak A,
    3. Bugarcic ZD
    : Platinum, palladium, gold and ruthenium complexes as anticancer agents: current clinical uses, cytotoxicity studies and future perspectives. Eur J Med Chem 142: 8-31, 2017.
    OpenUrl
  12. ↵
    1. Alessio E
    : Thirty years of the drug candidate NAMI-A and the myths in the field of ruthenium anticancer compounds: A personal perspective. Eur J Inorg Chem pp. 1549-1560, 2017.
  13. ↵
    1. Thota S,
    2. Rodrigues DA,
    3. Crans DC,
    4. Barreiro EJ
    : Ru(II) compounds: Next-generation anticancer metallotherapeutics? J Med Chem 61: 5805-5821, 2018.
    OpenUrl
  14. ↵
    1. Romero-Canelon I,
    2. Sadler PJ
    : Next-generation metal anticancer complexes: Multitargeting via redox modulation. Inorg Chem 52: 12276-12291, 2013.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Liu S,
    2. Cao W,
    3. Yu L,
    4. Zheng W,
    5. Li L,
    6. Fan C,
    7. Chen T
    : Zinc(II) complexes containing bis-benzimidazole derivatives as a new class of apoptosis inducers that trigger DNA damage-mediated p53 phosphorylation in cancer cells. Dalton Trans 42: 5932-5940, 2013.
    OpenUrl
    1. Hu J,
    2. Guo Y,
    3. Zhao J,
    4. Zhang J
    : In vitro antitumor activity of novel benzimidazole-based Cu(II) complexes. Bioorg Med Chem 25: 5733-5742, 2017.
    OpenUrl
  16. ↵
    1. Hussain A,
    2. AlAjmi MF,
    3. Rehman MT,
    4. Khan AA,
    5. Shaikh PA,
    6. Khan RA
    : Evaluation of transition metal complexes of benzimidazole-derived scaffold as promising anticancer chemotherapeutics. Molecules 23: 1232, 2018.
    OpenUrl
  17. ↵
    1. Demmer CS,
    2. Bunch L
    : Benzoxazoles and oxazolopyridines in medicinal chemistry studies. Eur J Med Chem 97: 778-785, 2015.
    OpenUrl
  18. ↵
    1. Kakkar S,
    2. Tahlan S,
    3. Lim SM,
    4. Ramasamy K,
    5. Mani V,
    6. Shah SAA,
    7. Narasimhan B
    : Benzoxazole derivatives: design, synthesis and biological evaluation. Chem Cent J 12: 92, 2018.
    OpenUrl
  19. ↵
    1. Kumar D,
    2. Jacob MR,
    3. Reynolds MB,
    4. Kerwin SM
    : Synthesis and evaluation of anticancer benzoxazoles and benzimidazoles related to UK-1. Bioorg Med Chem 10: 3997-4004, 2002.
    OpenUrlPubMed
    1. Rida SM,
    2. Ashour FA,
    3. El-Hawash SA,
    4. ElSemary MM,
    5. Badr MH,
    6. Shalaby MA
    : Synthesis of some novel benzoxazole derivatives as anticancer, anti-HIV-1 and antimicrobial agents. Eur J med Chem 40: 949-959, 2005.
    OpenUrlPubMed
    1. McKee ML,
    2. Kerwin SM
    : Synthesis, metal ion binding, and biological evaluation of new anticancer 2-(2’-hydroxyphenyl) benzoxazole analogs of UK-1. Bioorg Med Chem 16: 1775-1783, 2008.
    OpenUrlPubMed
    1. Duroux R,
    2. Agouridas L,
    3. Renault N,
    4. Bakali JEI,
    5. Furman C,
    6. Melnyk P,
    7. Yous S
    : Antagonists of the adenosine A2A receptor based on a 2-arylbenzoxazole scaffold: Investigation of the C5-and C7-positions to enhance affinity. Eur J med Chem 144: 151-163, 2018.
    OpenUrl
  20. ↵
    1. Zhang H-Z,
    2. Zhao Z-L,
    3. Zhou C-H
    : Recent advance in oxazole-based medicinal chemistry. Eur J Med Chem 144: 444-492, 2018.
    OpenUrl
  21. ↵
    1. Watanabe G,
    2. Sekiya H,
    3. Tamai E,
    4. Saijo R,
    5. Uno H,
    6. Mori S,
    7. Tanaka T,
    8. Maki J,
    9. Kawase M
    : Synthesis and antimicrobial activity of 2-trifluoroacetonylbenzoxazole ligands and their metal complexes. Chem Pharm Bull 66: 732-740, 2018.
    OpenUrl
  22. ↵
    1. Gupta RK,
    2. Sharma G,
    3. Pandey RA
    : DNA/protein binding, molecular docking, and in vitro anticancer activity of some thioether-dipyrrinato complexes. Inorg Chem 52: 13984-13996, 2013.
    OpenUrl
  23. ↵
    1. Hu J,
    2. Guo Y,
    3. Zhao J,
    4. Zhang J
    : In vitro antitumor activity of novel benzimidazole-based Cu(II) complexes. J Bioorg Med Chem 25: 5733-5742, 2017.
    OpenUrl
  24. ↵
    1. Mucsi I,
    2. Varga A,
    3. Kawase M,
    4. Motohashi N,
    5. Molnar J
    : Interaction between various resistance modifiers and apoptosis inducer 12H-benzo[a]phenothiazine. Anticancer Res 22: 2833-2836, 2002.
    OpenUrlPubMed
  25. ↵
    1. Tsuruo T,
    2. Iida H,
    3. Tsukagoshi S,
    4. Sakurai Y,
    5. Tsuruo T
    : Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil. Cancer Res 41: 1967-1972, 1981.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Voigt B,
    2. Coburger C,
    3. Monár J,
    4. Hilgeroth A
    : Structure–activity relationships of novel N-acyloxy-1,4-dihydropyridines as P-glycoprotein inhibitors. Bioorg Med Chem 15: 5110-5113, 2007.
    OpenUrlPubMed
  27. ↵
    1. Vock CA,
    2. Ang WH,
    3. Scolaro C,
    4. Phillips AD,
    5. Lagopoulos L,
    6. Juillerat-Jeanneret L,
    7. Sava G,
    8. Scopelliti R,
    9. Dyson PJ
    : Development of ruthenium antitumor drugs that overcome multidrug resistance mechanisms. J Med Chem 50: 2166-2175, 2007.
    OpenUrlPubMed
  28. ↵
    1. Kawase M,
    2. Sakagami H,
    3. Kusama K,
    4. Motohashi N,
    5. Saito S
    : α Trifluoromethylated acyloins induce apoptosis in human oral tumor cell lines. Bioorg Med Chem Lett 9: 3113-3118, 1999.
    OpenUrlPubMed
  29. ↵
    1. Sushama PG,
    2. Dorothy AM,
    3. Alaudeen M
    : Antimicrobial and antifungal activity of Mn(III) complexes of 2,5-dihydroxy-3-undecyl-1,4-benzoquinone derivative. Asian J Chem 19: 3403-3406, 2007.
    OpenUrl
    1. Li S,
    2. Hu P,
    3. Malmstadt N
    : Confocal imaging to quantify passive transport across biomimetic lipid membranes. Anal Chem 82: 7766-7771, 2010.
    OpenUrlPubMed
    1. Hunoor RS,
    2. Patil BR,
    3. Badiger DS,
    4. Vadavi RS,
    5. Gudasi KB
    : 2D HETCOR studies of 1,2-dihydroquinazolinone derivative: Synthesis characterization and anti-microbial study of its transition metal complexes. Pharm Chem 2: 116-128, 2010.
    OpenUrl
    1. Aravindhan R,
    2. Sreelatha T,
    3. Perumal PT,
    4. Gnanamani A
    : Synthesis, characterization and biological profile of metal and azo-metal complexes of embelin. Complex Metals 1: 69-79, 2014.
    OpenUrl
  30. ↵
    1. El-Tabl AS,
    2. Abd-El Wahed MM,
    3. Abu-Setta MHH
    : Metallo-bioactive compounds as potential novel anticancer therapy. Int J Adv Chem (IJAC) 4: 17-37, 2018.
    OpenUrl
  31. ↵
    1. Wu C-P,
    2. Calcagno AM,
    3. Ambudkar SV
    : Reversal of ABC drug transporter-mediated multidrug resistance in cancer cells: Evaluation of current strategies. Curr Mol Pharmacol 1: 93-105, 2008.
    OpenUrlCrossRefPubMed
    1. Beretta GL,
    2. Cassinelli G,
    3. Pennati M,
    4. Zuco V,
    5. Gatti L
    : Overcoming ABC transporter-mediated multidrug resistance: The dual role of tyrosine kinase inhibitors as multitargeting agents. Eur J Med Chem 142: 271-289, 2017.
    OpenUrl
  32. ↵
    1. Jaramillo AC,
    2. Saig FA,
    3. Cloos1 J,
    4. Jansen G,
    5. Peters GJ
    : How to overcome ATP-binding cassette drug efflux transporter-mediated drug resistance? Cancer Drug Resist 1: 6-29, 2018.
    OpenUrl
  33. ↵
    1. Jaszczyszyn A,
    2. Gasiorowski K,
    3. Swiatek P,
    4. Malinka W,
    5. Cieslik-Boczula K,
    6. Petrus J,
    7. Czzarnik-Matusewicz C
    : Chemical structure of phenothiazines and their biological activity. Pharmacol Rep 64: 16-23, 2012.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 38 (11)
Anticancer Research
Vol. 38, Issue 11
November 2018
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Benzoxazole-based Zn(II) and Cu(II) Complexes Overcome Multidrug-resistance in Cancer
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
5 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Benzoxazole-based Zn(II) and Cu(II) Complexes Overcome Multidrug-resistance in Cancer
GABRIELLA SPENGLER, ANNAMÁRIA KINCSES, BÁLINT RÁCZ, BORISZ VARGA, GENKI WATANABE, RYOSUKE SAIJO, HIROSHI SEKIYA, EIJI TAMAI, JUN MAKI, JOSEPH MOLNÁR, MASAMI KAWASE
Anticancer Research Nov 2018, 38 (11) 6181-6187; DOI: 10.21873/anticanres.12971

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Benzoxazole-based Zn(II) and Cu(II) Complexes Overcome Multidrug-resistance in Cancer
GABRIELLA SPENGLER, ANNAMÁRIA KINCSES, BÁLINT RÁCZ, BORISZ VARGA, GENKI WATANABE, RYOSUKE SAIJO, HIROSHI SEKIYA, EIJI TAMAI, JUN MAKI, JOSEPH MOLNÁR, MASAMI KAWASE
Anticancer Research Nov 2018, 38 (11) 6181-6187; DOI: 10.21873/anticanres.12971
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Conclusion
    • Acknowledgements
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • New Chalcone Derivative Inhibits ABCB1 in Multidrug Resistant T-cell Lymphoma and Colon Adenocarcinoma Cells
  • Google Scholar

More in this TOC Section

  • SOX10 Inhibits T Cell Recognition by Inducing Expression of the Immune Checkpoint Molecule PD-L1 in A375 Melanoma Cells
  • The Application of Static Magnetic Stimulation Reduces Survival of SH-SY5Y Neuroblastoma Cells
  • The Prognostic Role of ACO2 in Renal Cell Carcinoma
Show more Experimental Studies

Similar Articles

Keywords

  • multidrug resistance
  • metal complex
  • benzoxazole
  • ABCB1
  • P-glycoprotein
  • mouse T-lymphoma
  • apoptosis
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire