Next Article in Journal
Quasicrystal Tilings in Three Dimensions and Their Empires
Previous Article in Journal
The Crystal Structure of Defect KBB’O6 Pyrochlores (B,B’: Nb,W,Sb,Te) Revisited from Neutron Diffraction Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Platinum Metal Complexes as Potential Anti-Triple Negative Breast Cancer Agents

by
Eva María Domínguez-Martís
1,
Diego Gabriel Mosteiro-Miguéns
2,
Lucía Vigo-Gendre
1,
David López-Ares
3,
Manuel Freire-Garabal
4,
María Jesús Núñez-Iglesias
4 and
Silvia Novío
4,*
1
Galician Public Health Care Service, Health Care Centre of Ordes, C/Ramón Ferreiro s/n, 15680 Ordes, A Coruña, Spain
2
Badalona Serveis Assistencials, Family and Community Nursing, Health Care Centre of Morera-Pomar, C/PERE III, Nº 1-7, 08915 Badalona, Barcelona, Spain
3
Galician Public Health Care Service, University Hospital Complex of A Coruña (CHUAC), C/Xubias de Arriba, 84, 15006 A Coruña, Spain
4
SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, C/San Francisco s/n, 15782 A Coruña, Spain
*
Author to whom correspondence should be addressed.
Crystals 2018, 8(10), 369; https://doi.org/10.3390/cryst8100369
Submission received: 12 August 2018 / Revised: 18 September 2018 / Accepted: 18 September 2018 / Published: 20 September 2018

Abstract

:
Breast cancer (BC) is the most common cancer in women worldwide, with a mortality rate that has been forecasted to rise in the next decade. This is especially worrying for people with triple-negative BC (TNBC), because of its unresponsiveness to current therapies. Different drugs to treat TNBC have been assessed, and, although platinum chemotherapy drugs seem to offer some hope, their drawbacks have motivated extensive investigations into alternative metal-based BC therapies. This paper aims to: (i) describe the preliminary in vitro and in vivo anticancer properties of non-platinum metal-based complexes (NPMBC) against TNBC; and (ii) analyze the likely molecular targets involved in their anticancer activity.

1. Introduction

Breast cancer (BC) is, despite therapeutic advances, the most common cancer among females (1.5 million women worldwide each year) and the leading cause of death from cancer in women (570,000 in 2015). Furthermore, its incidence is expected to rise in the coming decades [1]. This global increase in the BC burden is mainly attributed to exogenous factors such as reproductive and lifestyle factors; however, genetic risk factors such as mutations in breast cancer susceptibility gene 1 (BRCA1) and gene 2 (BRCA2), which are associated to tumors with more aggressive phenotypes, play an important role in the etiology of this type of cancer [2,3].
BC is classified according to the expression of immunohistochemical markers in luminal A, luminal B, HER-2 positive and triple negative subtypes, being the triple negative subtype further divided into A (luminal-like) and B (basal-like) [4,5] (Table 1). The basal-like cancers account for 60–90% of triple negative cases, with the BC subtype having the worst prognosis. Compared with the other subtypes, triple negative breast cancer (TNBC) is more likely to be diagnosed at a younger age; it is more frequently associated with mutations in the BRCA; it usually has rapid growth and large size; it frequently overexpresses genes involved in drug-desensitizing mechanisms (for example, ABCC1 or MRP1 (multidrug resistance protein 1)); and it has a high rate of early recurrence and of distant metastasis (brain, lung, bone, and liver), and a low disease-specific survival [6]. Furthermore, due to the lack of specific molecular targets in this subtype of BC [7], which makes its treatment difficult, chemotherapy remains an essential component for the management of TNBC, both adjuvant and neoadjuvant therapy [8,9].
Besides their diagnostic use [14,15,16,17,18,19,20,21], metallodrugs or metal-based complexes (MBC) stand out for being promising chemotherapeutic agents [22]. The therapeutic potential of MBC has long been known; however, their role in the treatment of cancer is relatively recent (1960s). The first metal that laid the foundations of the modern era of metal-based anticancer drugs was platinum, and particularly its drug-derived cisplatin of which cytotoxic activity depends on apoptotic mechanisms and DNA repair defects caused by BRCA1 dysfunction [23,24], hence BRCA-deficient TNBC cells are particularly sensitive to it [25]. However, both cisplatin chemotherapies and its analogs have been shown to have major drawbacks (i.e., intrinsic and acquired chemoresistance, high general toxicity and limited spectrum of activity) [26] which have motivated extensive investigations into alternative metal-based cancer therapies that effectively target both cancer cell proliferation and metastasis. In this sense, we can mention chemotherapy with gold compounds [27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46]—because they are nontoxic, nonimmunogenic and have good photothermal and optical properties, biocompatibility and stability [40,47,48]—and also with copper [49,50,51], ruthenium [16,52,53,54,55,56,57,58,59], iron [56,60,61,62,63,64,65], palladium [50], silver [66,67], iridium [68], osmium [68] and rhodium [69] compounds.
This paper aims to: (i) describe the preliminary in vitro and in vivo anticancer properties of non-platinum MBC (NPMBC) against TNBC; and (ii) analyze the likely molecular targets involved in their anticancer activity.

2. Anticancer Effects of Non-Platinum MBC (NPMBC)

The mechanisms behind the anticancer effects of NPMBC are not fully understood, but known responses to them include the following: (i) suppression of cancer cell viability (Table 2) in association with induction of cell death (Table 3 and Table 4); and (ii) inhibition of metastatic processes (Table 5). Furthermore, a study suggests that NPMBC could also inhibit angiogenesis [55]; however, because of scarce evidence, no definitive conclusion can be drawn on the role of these compounds in the regulation of blood vessel formation. In any case, it is thought that NPMBC are frequently multi-targeted and attack different biochemical pathways simultaneously [53,66,68], sometimes in a synergic way [37,62].
Cellular studies constitute an important step in the development of drugs; however, demonstrating efficacy in suitable animal models and validating cellular observations in vivo are a condition sine qua non for clinical trials investigating potential anticancer agents. Inconsistencies in results between in vitro and in vivo systems concerning the anticancer effect of NPMBC can be related to differences in their pharmacokinetic behavior, thus their study is necessary. In this way, NPMBC seem to have good in vivo pharmacokinetic properties. They are absorbed quickly into plasma [16,39,44,52], the circulation time in the bloodstream is long [42], their uptake in the tumor tissue is significant [16,28,32,37,39,41,42,44,46,51,52,60,70], their accumulation in organs (brain, liver, kidney, spleen, heart, lung, and intestine) is low [16,28,39,41,42,44,51,52,55] and the clearing rate is good [51].
A large proportion of current knowledge about the therapeutic potential of NPMBC is derived from in vitro and in vivo studies performed using TNBC cell lines. TNBC MDA-MB-231 cell models are considered the most reliable models of TNBC [71,72], so this cell line is the world’s most commonly used human TNBC cell line. However, this raises the issue on how representative this cell line is of the broad spectrum of TNBCs (Table 1).

2.1. Suppression of Cancer Cell Viability in Association with the Induction of Cell Death

NPMBC-mediated inhibition of breast carcinogenesis is associated with reduced cellular proliferation (Table 2) together with the induction of programmed cell death (Table 3 and Table 4). Apoptosis is by far the primary mode of programmed cell death (Table 3), but sometimes it is coupled to the induction of autophagy, as can be suspected from the few studies which have been carried out (Table 4).
NPMBC display good cytotoxic behavior in vitro and are able to reduce the growth of primary tumors (Table 2). NPMBC inhibit the growth of TNBC cells better than non-TNBC cells [66]. Furthermore, NPMBC of which effects are dose- [49,53,55,67] and time- [63,67] dependent could be more cytotoxic than platinum-based drugs [52,53,57,58,68]. For example, Biancalana et al. [57] observed that ruthenium complexes have good cytotoxic activity, with IC50 values substantially lower than the values obtained with cisplatin on MDA-MB-231 cells. These differences could be due to the fact that they have different mechanisms of action; for example, the activation of p53-dependent or p53-independent checkpoints by cisplatin or ruthenium complexes, respectively [9,52,54,68]. Since platinum-based drugs and NPMBC act on different pathways, NPMBC could be a new therapeutic option for patients with TNBC resistant to platinum-based drugs.
Avoiding apoptosis is a hallmark of cancer and an important mechanism in resistance to therapies. Apoptosis is preferred over necrosis because the latter elicits inflammation and undesirable immunogenic responses [73]. NPMBC induce significant apoptotic activity in TNBC cells which can be mediated by multiple mechanisms (Table 3). When MDA-MB-231 and BT-20 cells were treated with 25 μM copper salicylate phenanthroline complexes, more than 80% of TNBC cells underwent apoptosis through down-regulation of the anti-apoptosis proteins Bcl-2, Bcl-xL and survivin and up-regulation of cleaved PARP [49]. These findings confirm the participation of protease enzyme caspases in the programmed cell death induced by NPMBC. However, NPMBC can also activate caspase-independent pathways acting on mitochondria. For example, when MDA-MB-231 cells were treated with 10 μg/mL of silver nanoparticles, high intracellular levels of reactive oxygen species (ROS) were observed at 24 h of incubation [67]. These free radicals destabilize the mitochondria and induce apoptosis [74] acting on Bcl-2 family proteins [75]. Considering that TNBC is more susceptible to drugs that cause oxidative stress than other BC types [66], these compounds could be a hopeful alternative method for its treatment. Besides what was mentioned above, we must not forget that the efficacy of several NPMBC relies on their capacity to influence the tumor–host interaction and modify the immune microenvironment, inducing a form of apoptosis in cancer cells known as “immunogenic cell death”. Thus, Montani et al. [55] observed that ruthenium complexes reverse tumor-associated immune suppression leading to the activation of an immune response specific for TNBC cells.
The NPMBC can be synthesized by chemical [76], physical [77] and biological methods [35], all with cytotoxic activity against TNBC cells. Although chemical and physical syntheses have been the most widely used strategies, there is growing interest in the biosynthesis of NPMBC because of its numerous advantages (for example, economical, safe, and inert) [78,79].
The NPMBC differ in shape [34], size [31,32,33,42], dose [30,36,66], synthesis procedure (see previous paragraph), composition [50,56], capping agent [33,80], particle surface charges [31] and structure [56,68,69]. All of these features can affect their cellular uptake and therefore also their cytotoxic activity. In general, modified small nanoparticles with low surface charges (−15 mV) have better cellular internalization than large nanoparticles, however both extremely small (<25 nm) and large (>80 nm) nanoparticles displayed a low cellular uptake [31,33]. On the other hand, it has been demonstrated that replacing ruthenium with iron gives compounds with high antiproliferative activity (IC50 = 0.09 vs. >30 μM), the compounds with the shortest carbon chain linking the two cyclopentadienyl rings being the most active [56]. Finally, another interesting finding is the superiority of nanorods over nanospheres for photothermal cancer therapy (PCT) using gold nanoparticles [34]. Despite this evidence, it has not been possible to establish a linear correlation between cellular responses and the characteristics of NPMBC because there is wide interstudy variability (for example, in relation to cell lines, measurement methods, etc.).
NPMBC possess cytotoxic activity per se [39,66,67,69]; however, their main applications are related to their ability to act as radiosensitizers in cancer cells through the induction of G2/M phase cell cycle arrest [36,47,66], as agents for PCT applications [14,28,29,37,63] or as transmembrane carriers for the controlled release and targeted delivery of anticancer drugs increasing their cellular uptake [30,39,40], among others [47,64]. Cancer cells in a tumor are very heterogeneous, that is, they differ in marker expression, morphology, proliferation capacity, growth stage, etc. This heterogeneity increases the need for combining therapeutic agents with different action mechanisms. Although few studies have examined the role of NPMBC for combination chemotherapy, recently the utility of iron oxide magnetic nanoparticles as multidrug codelivery system for synergistic chemotherapy has been demonstrated [63].
The effects of NPMBC on TNBC cells, as well as on cancer cells from other solid tumors, is facilitated by abnormalities of the tumor vasculature, such as hypervascularization, high production of vascular permeability factors, etc. [81]. However, intrinsic properties of NPMBC are not less important. For example, nanoparticles are characterized by their high permeability [82] and retention effect [83] which results in potentiation of the cytotoxic effects with minimal side effects [39,49]. When the tyrosine kinase inhibitor ZD6474 was conjugated with gold nanoparticles, besides inducing a higher antiproliferative effect than the ZD6474 alone under in vitro conditions, it also reduced the size of tumors induced in mice more efficiently than the drug alone [44]. These properties could be useful to counteract drug resistance mechanisms, the main limitation to the success of conventional therapy. In this sense, Conde et al. [41] showed that gold nanoparticles loaded with 5-fluorouracil (IC50 = ~0.5 nM) decrease the viability of 5-fluorouracil-resistant MDA-MB-231 cells by silencing the cell surface efflux pump MRP1.
One of the concerns for developing NPMBC as therapeutic agents for TNBC treatment could be their toxicity. However, several studies have proven their harmlessness in normal cells [39,49,52,57,66,84]. Ruthenium(II) paracyme complexes showed a moderate selectivity for TNBC cells, so normal cells (human skin fibroblasts) were less affected than MDA-MB-231 cells [57]. Likewise, when copper salicylate phenanthroline complexes [49], ruthenium complexes [55] or gold nanoparticles [28,37,39] were administered to mice, they were well tolerated as evidenced by the lack of weight loss, behavior of the animal or examination of organs.
It is hypothesized that, because of their capacity for self-renewal, differentiation and apoptosis-resistance [85], cancer stem cells (CSCs, also referred to as tumor-initiating cells) could be responsible for the cancer formation [61]. What is more, they could be resistant to conventional treatments and contribute to the recurrence after treatment [86]. Gold nanoshells seem to sensitize breast CSCs to radiation therapy mediated by the activation of the heat shock proteins (HSP) HSP40, HSP60, HSP70 and HSP90α [45]. Likewise, CSCs’ ability to self-renew might be inhibited by iron oxide nanoparticle-mediated PCT [61] or iron oxide nanoparticles-linked leptin antagonist [64] which has been demonstrated through the reduction of mammospheres/tumorspheres of MDA-MB-231 and SUM 159 cells [61,64]. Although these findings are hopeful, new strategies for improving the intratumoral distribution of NPMBC are necessary to achieve a homogeneous nanoparticle distribution within the tumor without depositing them in surrounding healthy tissue [70].

2.2. Migration, Invasion and/or Metastasis

Metastasis is a cascade of events where tumor cells disseminate from the primary tumor to distant sites, with the migration of tumor cells being a prerequisite for tumor-cell invasion and metastasis (Figure 1) [87]. Thus, any factor which regulates this process might be a target for anti-metastatic therapeutic strategies (Table 5). The poor prognosis of the triple negative subtype reflects the aggressive biology of this disease and the difficulty of its treatment, especially when it metastasizes.
Study of the anti-metastatic effect of NPMBC on TNBC cells is very recent. It has been shown that NPMBC, besides enhancing the potential of anticancer therapies [43,44,62], can be used as monotherapy [43,51] to prevent cancer cell migration, invasion and/or metastasis. These findings were confirmed by Paholak et al. [61] using in vivo models. When immunocompromised NOD/SCID mice were inoculated with TNBC cells, it was observed that those mice that received nanoparticle-mediated PCT prior to surgical tumor resection had lower incidence of metastasis to the lung than those which had been treated only with surgery. If these findings were observed in humans, combination therapy consisting of nanoparticle-mediated PCT and surgery would provide a treatment option for patients with metastatic breast cancer, a disease which nowadays has no cure [88].
Unfortunately, little is known about the mechanisms underlying the anti-metastatic effect of NPMBC (Figure 1 and Table 5). However, available evidence suggests that their action could be mediated by the inhibition of matrix metalloproteinases (MMPs) [59], enzymes with the ability to degrade extracellular matrix proteins, after interaction with the αvβ3 integrin receptor [62]. Likewise, it has been suggested that NPMBC could inhibit epithelial–mesenchymal transition [65] and modify the structure of the actin cytoskeleton, interfering with the function of integrins [58,59] and under-regulating the phosphorylation of the focal adhesion kinase (FAK) [51]. No less important are the studies which highlight their role in inflammation [89,90]. On the one hand, it has been observed that doxorubicin-hyaluronan conjugated iron oxide nanoparticles reduce the secretion of the pro-inflammatory IL-6, an important prognostic indicator in patients with TNCB [91], and which is thought to contribute to their invasiveness [92]; on the other hand, it has been suggested that iron oxide nanoparticle-mediated hyperthermia could trigger the release of inflammatory mediators which would induce a systemic cancer-specific immune response in which cytotoxic T-cells would recognize and inhibit distal cancer cells [89].
One of the major limitations of anti-metastatic chemotherapy, as has already been mentioned for the treatment of primary tumors, is the toxicity which underlies the importance of targeted therapies. Anticancer drugs should be delivered and act only at the site of action. In this respect, nanoparticles are worthy of mention because they more effectively inhibit the migration of TNBC cells compared to non-tumor cells [51]. Sarkar et al. [44] found potentiation of the anti-migration and anti-invasion effect of the tyrosine kinase inhibitor ZD6474 when it was conjugated to gold nanoparticles synthesized in micellar networks of an amphiphilic block copolymer. On the other hand, the conjugation of interfering RNA (siRNA) to nanoparticles in order to suppress the expression of proteins is another interesting approach. It has been shown that chitosan-layered gold nanorods can be used for silencing genes related to the invasion of TNBC cells [43]. Another strategy which enables the selectivity for tumor cells to be enhanced is by attaching nanoparticles to ligands that bind to receptors expressed on the surface of cancer cells, as has been highlighted by Ahir et al. [51]. Considering that folate receptor expression is elevated in BC cells [93], this receptor could be a good target for cancer treatment. Copper oxide nanowires conjugated with folic acid prevent the motility of MDA-MB-231 cells, an effect that has been validated in vivo [51].

3. Conclusions

Anticancer drugs with new molecular mechanisms of action are necessary for chemotherapy treatment of TNBC, with NPMBC emerging as an upcoming treatment modality. It has been suggested that NPMBC could have an effect on different TNBC cell lines, both in vitro and in vivo. In this way, it has been shown that NPMBC are responsible for activating programmed cell death and exhibiting activity against metastasis. Despite these findings, more studies with different cell lines are required to explore other pathways which could contribute to the control of the broad spectrum of TNBC.

Author Contributions

E.M.D.M. contributed substantially to the conception of the work, analysis of data, drafting, critical revision and supervision of the submitted manuscript. D.G.M.-M. contributed substantially to the conception of the work, analysis of data, drafting and critical revision of the submitted manuscript. L.V.-G. and D.L.-A. contributed substantially to the design of the work, acquisition and analysis of data, and drafting of the submitted manuscript. M.J.N.-I. contributed substantially to the design of the work, acquisition, analysis and interpretation of data, and drafting of the submitted manuscript. M.F.-G. contributed substantially to the conception of the work, interpretation of data and critical revision of the submitted manuscript. S.N. contributed substantially to the analysis and interpretation of data, design, drafting, critical revision and supervision of the submitted manuscript. Furthermore, all authors have approved the final version to be published and are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated, resolved and documented in the literature.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Breast Cancer. Available online: http://www.who.int/cancer/prevention/diagnosis-screening/breast-cancer/en/ (accessed on 27 July 2018).
  2. Howell, A.; Anderson, A.S.; Clarke, R.B.; Duffy, S.W.; Evans, D.G.; Garcia-Closas, M.; Gescher, A.J.; Key, T.J.; Saxton, J.M.; Harvie, M.N. Risk Determination and Prevention of Breast Cancer. Breast Cancer Res. 2014, 16, 446. [Google Scholar] [CrossRef] [PubMed]
  3. Ricks-Santi, L.; McDonald, J.T.; Gold, B.; Dean, M.; Thompson, N.; Abbas, M.; Wilson, B.; Kanaan, Y.; Naab, T.J.; Dunston, G. Next Generation Sequencing Reveals High Prevalence of BRCA1 and BRCA2 Variants of Unknown Significance in Early-Onset Breast Cancer in African American Women. Ethn. Dis. 2017, 27, 169–178. [Google Scholar] [CrossRef] [PubMed]
  4. Dai, X.; Cheng, H.; Bai, Z.; Li, J. Breast Cancer Cell Line Classification and Its Relevance with Breast Tumor Subtyping. J. Cancer 2017, 8, 3131–3141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Zaha, D.C. Significance of Immunohistochemistry in Breast Cancer. World J. Clin. Oncol. 2014, 5, 382–392. [Google Scholar] [CrossRef] [PubMed]
  6. Aysola, K.; Desai, A.; Welch, C.; Xu, J.; Qin, Y.; Reddy, V.; Matthews, R.; Owens, C.; Okoli, J.; Beech, D.J.; et al. Triple Negative Breast Cancer—An Overview. Hered. Genet. 2013, 2013. [Google Scholar] [CrossRef]
  7. Sharma, P. Update on the Treatment of Early-Stage Triple-Negative Breast Cancer. Curr. Treat. Options Oncol. 2018, 19, 22. [Google Scholar] [CrossRef] [PubMed]
  8. Miller, E.; Lee, H.J.; Lulla, A.; Hernandez, L.; Gokare, P.; Lim, B. Current Treatment of Early Breast Cancer: Adjuvant and Neoadjuvant Therapy. F1000Res 2014, 3, 198. [Google Scholar] [CrossRef] [PubMed]
  9. Hongthong, K.; Ratanaphan, A. BRCA1-Associated Triple-Negative Breast Cancer and Potential Treatment for Ruthenium-Based Compounds. Curr. Cancer Drug Targets 2016, 16, 606–617. [Google Scholar] [CrossRef] [PubMed]
  10. Chavez, K.J.; Garimella, S.V.; Lipkowitz, S. Triple Negative Breast Cancer Cell Lines: One Tool in the Search for Better Treatment of Triple Negative Breast Cancer. Breast Dis. 2010, 32, 35–48. [Google Scholar] [CrossRef] [PubMed]
  11. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [PubMed]
  12. Neve, R.M.; Chin, K.; Fridlyand, J.; Yeh, J.; Baehner, F.L.; Fevr, T.; Clark, L.; Bayani, N.; Coppe, J.P.; Tong, F.; et al. A Collection of Breast Cancer Cell Lines for the Study of Functionally Distinct Cancer Subtypes. Cancer Cell 2006, 10, 515–527. [Google Scholar] [CrossRef] [PubMed]
  13. Jiang, G.; Zhang, S.; Yazdanparast, A.; Li, M.; Pawar, A.V.; Liu, Y.; Inavolu, S.M.; Cheng, L. Comprehensive Comparison of Molecular Portraits Between Cell Lines and Tumors in Breast Cancer. BMC Genomics 2016, 17, 525. [Google Scholar] [CrossRef] [PubMed]
  14. Webb, J.A.; Ou, Y.C.; Faley, S.; Paul, E.P.; Hittinger, J.P.; Cutright, C.C.; Lin, E.C.; Bellan, L.M.; Bardhan, R. Theranostic Gold Nanoantennas for Simultaneous Multiplexed Raman Imaging of Immunomarkers and Photothermal Therapy. ACS Omega 2017, 2, 3583–3594. [Google Scholar] [CrossRef] [PubMed]
  15. Tao, Y.; Li, M.; Auguste, D.T. Pattern-Based Sensing of Triple Negative Breast Cancer Cells with Dual-Ligand Cofunctionalized Gold Nanoclusters. Biomaterials 2017, 116, 21–33. [Google Scholar] [CrossRef] [PubMed]
  16. Shen, J.; Kim, H.C.; Wolfram, J.; Mu, C.; Zhang, W.; Liu, H.; Xie, Y.; Mai, J.; Zhang, H.; Li, Z.; et al. A Liposome Encapsulated Ruthenium Polypyridine Complex as a Theranostic Platform for Triple-Negative Breast Cancer. Nano Lett. 2017, 17, 2913–2920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Zhao, Y.; Pang, B.; Luehmann, H.; Detering, L.; Yang, X.; Sultan, D.; Harpstrite, S.; Sharma, V.; Cutler, C.S.; Xia, Y.; et al. Gold Nanoparticles Doped with (199) Au Atoms and Their Use for Targeted Cancer Imaging by SPECT. Adv. Healthc. Mater. 2016, 5, 928–935. [Google Scholar] [CrossRef] [PubMed]
  18. Zhang, M.; Kim, H.S.; Jin, T.; Yi, A.; Moon, W.K. Ultrasound-Guided Photoacoustic Imaging for the Selective Detection of EGFR-Expressing Breast Cancer and Lymph Node Metastases. Biomed. Opt. Express 2016, 7, 1920–1931. [Google Scholar] [CrossRef] [PubMed]
  19. Abulrob, A.; Corluka, S.; Blasiak, B.; Gino Fallone, B.; Ponjevic, D.; Matyas, J.; Tomanek, B. LyP-1 Conjugated Nanoparticles for Magnetic Resonance Imaging of Triple Negative Breast Cancer. Mol. Imaging Biol. 2018, 20, 428–435. [Google Scholar] [CrossRef] [PubMed]
  20. Guo, P.; Huang, J.; Wang, L.; Jia, D.; Yang, J.; Dillon, D.A.; Zurakowski, D.; Mao, H.; Moses, M.A.; Auguste, D.T. ICAM-1 as a Molecular Target for Triple Negative Breast Cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 14710–14715. [Google Scholar] [CrossRef] [PubMed]
  21. Pang, B.; Zhao, Y.; Luehmann, H.; Yang, X.; Detering, L.; You, M.; Zhang, C.; Zhang, L.; Li, Z.Y.; Ren, Q.; et al. 64Cu-Doped PdCu@Au Tripods: A Multifunctional Nanomaterial for Positron Emission Tomography and Image-Guided Photothermal Cancer Treatment. ACS Nano 2016, 10, 3121–3131. [Google Scholar] [CrossRef] [PubMed]
  22. Ndagi, U.; Mhlongo, N.; Soliman, M.E. Metal Complexes in Cancer Therapy—An Update from Drug Design Perspective. Drug Des. Dev. Ther. 2017, 11, 599–616. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, D.; Lippard, S.J. Cellular Processing of Platinum Anticancer Drugs. Nat. Rev. Drug Discov. 2005, 4, 307–320. [Google Scholar] [CrossRef] [PubMed]
  24. Birkbak, N.J.; Wang, Z.C.; Kim, J.Y.; Eklund, A.C.; Li, Q.; Tian, R.; Bowman-Colin, C.; Li, Y.; Greene-Colozzi, A.; Iglehart, J.D.; et al. Telomeric Allelic Imbalance Indicates Defective DNA Repair and Sensitivity to DNA-Damaging Agents. Cancer Discov. 2012, 2, 366–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Silver, D.P.; Richardson, A.L.; Eklund, A.C.; Wang, Z.C.; Szallasi, Z.; Li, Q.; Juul, N.; Leong, C.O.; Calogrias, D.; Buraimoh, A.; et al. Efficacy of Neoadjuvant Cisplatin in Triple-Negative Breast Cancer. J. Clin. Oncol. 2010, 28, 1145–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ciarimboli, G. Membrane Transporters as Mediators of Cisplatin Side-Effects. Anticancer Res. 2014, 34, 547–550. [Google Scholar] [CrossRef] [PubMed]
  27. Dam, D.H.; Culver, K.S.; Odom, T.W. Grafting Aptamers onto Gold Nanostars Increases in vitro Efficacy in a Wide Range of Cancer Cell Types. Mol. Pharm. 2014, 11, 580–587. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, S.; Tian, Y.; Tian, W.; Sun, J.; Zhao, S.; Liu, Y.; Wang, C.; Tang, Y.; Ma, X.; Teng, Z.; et al. Selectively Sensitizing Malignant Cells to Photothermal Therapy Using a CD44-Targeting Heat Shock Protein 72 Depletion Nanosystem. ACS Nano 2016, 10, 8578–8590. [Google Scholar] [CrossRef] [PubMed]
  29. Ou, Y.C.; Webb, J.A.; Faley, S.; Shae, D.; Talbert, E.M.; Lin, S.; Cutright, C.C.; Wilson, J.T.; Bellan, L.M.; Bardhan, R. Gold Nanoantenna-Mediated Photothermal Drug Delivery from Thermosensitive Liposomes in Breast Cancer. ACS Omega 2016, 1, 234–243. [Google Scholar] [CrossRef] [PubMed]
  30. Santiago, T.; DeVaux, R.S.; Kurzatkowska, K.; Espinal, R.; Herschkowitz, J.I.; Hepel, M. Surface-Enhanced Raman Scattering Investigation of Targeted Delivery and Controlled Release of Gemcitabine. Int. J. Nanomed. 2017, 12, 7763–7776. [Google Scholar] [CrossRef] [PubMed]
  31. Saw, W.S.; Ujihara, M.; Chong, W.Y.; Voon, S.H.; Imae, T.; Kiew, L.V.; Lee, H.B.; Sim, K.S.; Chung, L.Y. Size-Dependent Effect of Cystine/Citric Acid-Capped Confeito-Like Gold Nanoparticles on Cellular Uptake and Photothermal Cancer Therapy. Colloids Surf. B Biointerfaces 2018, 161, 365–374. [Google Scholar] [CrossRef] [PubMed]
  32. Ayala-Orozco, C.; Urban, C.; Bishnoi, S.; Urban, A.; Charron, H.; Mitchell, T.; Shea, M.; Nanda, S.; Schiff, R.; Halas, N.; et al. Sub-100 nm Gold Nanomatryoshkas Improve Photo-Thermal Therapy Efficacy in Large and Highly Aggressive Triple Negative Breast Tumors. J. Control. Release 2014, 191, 90–97. [Google Scholar] [CrossRef] [PubMed]
  33. Kong, T.; Zeng, J.; Wang, X.; Yang, X.; Yang, J.; McQuarrie, S.; McEwan, A.; Roa, W.; Chen, J.; Xing, J.Z. Enhancement of Radiation Cytotoxicity in Breast-Cancer Cells by Localized Attachment of Gold Nanoparticles. Small 2008, 4, 1537–1543. [Google Scholar] [CrossRef] [PubMed]
  34. Dube, E.; Oluwole, D.O.; Nwaji, N.; Nyokong, T. Glycosylated Zinc Phthalocyanine-Gold Nanoparticle Conjugates for Photodynamic Therapy: Effect of Nanoparticle Shape. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2018, 203, 85–95. [Google Scholar] [CrossRef] [PubMed]
  35. Dozie-Nwachukwu, S.O.; Obayemi, J.D.; Danyuo, Y.; Anuku, N.; Odusanya, O.S.; Malatesta, K.; Soboyejo, W.O. A Comparative Study of the Adhesion of Biosynthesized Gold and Conjugated Gold/Prodigiosin Nanoparticles to Triple Negative Breast Cancer Cells. J. Mater. Sci. Mater. Med. 2017, 28, 143. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, C.; Jiang, Y.; Li, X.; Hu, L. Thioglucose-Bound Gold Nanoparticles Increase the Radiosensitivity of a Triple-Negative Breast Cancer Cell Line (MDA-MB-231). Breast Cancer 2015, 22, 413–420. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, M.; Kim, H.S.; Jin, T.; Woo, J.; Piao, Y.J.; Moon, W.K. Near-Infrared Photothermal Therapy Using Anti-EGFR-Gold Nanorod Conjugates for Triple Negative Breast Cancer. Oncotarget 2017, 8, 86566–86575. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, M.; Kim, H.S.; Jin, T.; Moon, W.K. Near-Infrared Photothermal Therapy Using EGFR-Targeted Gold Nanoparticles Increases Autophagic Cell Death in Breast Cancer. J. Photochem. Photobiol. B. 2017, 170, 58–64. [Google Scholar] [CrossRef] [PubMed]
  39. Malik, M.T.; O’Toole, M.G.; Casson, L.K.; Thomas, S.D.; Bardi, G.T.; Reyes-Reyes, E.M.; Ng, C.K.; Kang, K.A.; Bates, P.J. AS1411-Conjugated Gold Nanospheres and Their Potential for Breast Cancer Therapy. Oncotarget 2015, 6, 22270–22281. [Google Scholar] [CrossRef] [PubMed]
  40. Haynes, B.; Zhang, Y.; Liu, F.; Li, J.; Petit, S.; Kothayer, H.; Bao, X.; Westwell, A.D.; Mao, G.; Shekhar, M.P.V. Gold Nanoparticle Conjugated Rad6 Inhibitor Induces Cell Death in Triple Negative Breast Cancer Cells by Inducing Mitochondrial Dysfunction and PARP-1 Hyperactivation: Synthesis and Characterization. Nanomedicine 2016, 12, 745–757. [Google Scholar] [CrossRef] [PubMed]
  41. Conde, J.; Oliva, N.; Artzi, N. Implantable Hydrogel Embedded Dark-Gold Nanoswitch as a Theranostic Probe to Sense and Overcome Cancer Multidrug Resistance. Proc. Natl. Acad. Sci. USA 2015, 112, E1278–E1287. [Google Scholar] [CrossRef] [PubMed]
  42. Ayala-Orozco, C.; Urban, C.; Knight, M.W.; Urban, A.S.; Neumann, O.; Bishnoi, S.W.; Mukherjee, S.; Goodman, A.M.; Charron, H.; Mitchell, T.; et al. Au Nanomatryoshkas as Efficient Near-Infrared Photothermal Transducers for Cancer Treatment: Benchmarking Against Nanoshells. ACS Nano 2014, 8, 6372–6381. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, Z.; Liu, T.; Xie, Y.; Sun, Z.; Liu, H.; Lin, J.; Liu, C.; Mao, Z.W.; Nie, S. Chitosan Layered Gold Nanorods as Synergistic Therapeutics for Photothermal Ablation and Gene Silencing in Triple-Negative Breast Cancer. Acta Biomater. 2015, 25, 194–204. [Google Scholar] [CrossRef] [PubMed]
  44. Sarkar, S.; Konar, S.; Prasad, P.N.; Rajput, S.; Kumar, B.N.P.; Rao, R.R.; Pathak, A.; Fisher, P.B.; Mandal, M. Micellear Gold Nanoparticles as Delivery Vehicles for Dual Tyrosine Kinase Inhibitor ZD6474 for Metastatic Breast Cancer Treatment. Langmuir 2017, 33, 7649–7659. [Google Scholar] [CrossRef] [PubMed]
  45. Atkinson, R.L.; Zhang, M.; Diagaradjane, P.; Peddibhotla, S.; Contreras, A.; Hilsenbeck, S.G.; Woodward, W.A.; Krishnan, S.; Chang, J.C.; Rosen, J.M. Thermal Enhancement with Optically Activated Gold Nanoshells Sensitizes Breast Cancer Stem Cells to Radiation Therapy. Sci. Transl. Med. 2010, 2. [Google Scholar] [CrossRef] [PubMed]
  46. Zhao, N.; Yang, Z.; Li, B.; Meng, J.; Shi, Z.; Li, P.; Fu, S. RGD-Conjugated Mesoporous Silica-Encapsulated Gold Nanorods Enhance the Sensitization of Triple-Negative Breast Cancer to Megavoltage Radiation Therapy. Int. J. Nanomed. 2016, 11, 5595–5610. [Google Scholar] [CrossRef] [PubMed]
  47. Lee, J.; Chatterjee, D.K.; Lee, M.H.; Krishnan, S. Gold Nanoparticles in Breast Cancer Treatment: Promise and Potential Pitfalls. Cancer Lett. 2014, 347, 46–53. [Google Scholar] [CrossRef] [PubMed]
  48. Klassen, N.V.; Kedrov, V.V.; Ossipyan, Y.A.; Shmurak, S.Z.; Shmyt Ko, I.M.; Krivko, O.A.; Kudrenko, E.A.; Kurlov, V.N.; Kobelev, N.P.; Kiselev, A.P.; et al. Nanoscintillators for Microscopic Diagnostics of Biological and Medical Objects and Medical Therapy. IEEE Trans. Nanobiosci. 2009, 8, 20–32. [Google Scholar] [CrossRef] [PubMed]
  49. Fan, L.; Tian, M.; Liu, Y.; Deng, Y.; Liao, Z.; Xu, J. Salicylate •Phenanthroline Copper (II) Complex Induces Apoptosis in Triple-Negative Breast Cancer Cells. Oncotarget 2017, 8, 29823–29832. [Google Scholar] [CrossRef] [PubMed]
  50. Afrasiabi, Z.; Stovall, P.; Finley, K.; Choudhury, A.; Barnes, C.; Ahmad, A.; Sarkar, F.; Vyas, A.; Padhye, S. Targeting Triple Negative Breast Cancer Cells by N3-Substituted 9,10-Phenanthrenequinone Thiosemicarbazones and Their Metal Complexes. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2013, 114, 114–119. [Google Scholar] [CrossRef] [PubMed]
  51. Ahir, M.; Bhattacharya, S.; Karmakar, S.; Mukhopadhyay, A.; Mukherjee, S.; Ghosh, S.; Chattopadhyay, S.; Patra, P.; Adhikary, A. Tailored-CuO-Nanowire Decorated with Folic Acid Mediated Coupling of the Mitochondrial-ROS Generation and miR425-PTEN Axis in Furnishing Potent Anti-Cancer Activity in Human Triple Negative Breast Carcinoma Cells. Biomaterials 2016, 76, 115–132. [Google Scholar] [CrossRef] [PubMed]
  52. Frik, M.; Martínez, A.; Elie, B.T.; Gonzalo, O.; Ramírez de Mingo, D.; Sanaú, M.; Sánchez-Delgado, R.; Sadhukha, T.; Prabha, S.; Ramos, J.W.; et al. In Vitro and in Vivo Evaluation of Water-Soluble Iminophosphorane Ruthenium(II) Compounds. A Potential Chemotherapeutic Agent for Triple Negative Breast Cancer. J. Med. Chem. 2014, 57, 9995–10012. [Google Scholar] [CrossRef] [PubMed]
  53. Irace, C.; Misso, G.; Capuozzo, A.; Piccolo, M.; Riccardi, C.; Luchini, A.; Caraglia, M.; Paduano, L.; Montesarchio, D.; Santamaria, R. Antiproliferative Effects of Ruthenium-Based Nucleolipidic Nanoaggregates in Human Models of Breast Cancer in Vitro: Insights into Their Mode of Action. Sci. Rep. 2017, 7, 45236. [Google Scholar] [CrossRef] [PubMed]
  54. Nhukeaw, T.; Temboot, P.; Hansongnern, K.; Ratanaphan, A. Cellular Responses of BRCA1-Defective and Triple-Negative Breast Cancer Cells and in Vitro BRCA1 Interactions Induced by Metallo-Intercalator Ruthenium(II) Complexes Containing Chloro-Substituted Phenylazopyridine. BMC Cancer 2014, 14, 73. [Google Scholar] [CrossRef] [PubMed]
  55. Montani, M.; Pazmay, G.V.B.; Hysi, A.; Lupidi, G.; Pettinari, R.; Gambini, V.; Tilio, M.; Marchetti, F.; Pettinari, C.; Ferraro, S.; et al. The Water Soluble Ruthenium(II) Organometallic Compound [Ru(p-cymene)(bis(3,5 dimethylpyrazol-1-yl)methane)Cl]Cl Suppresses Triple Negative Breast Cancer Growth by Inhibiting Tumor Infiltration of Regulatory T Cells. Pharmacol. Res. 2016, 107, 282–290. [Google Scholar] [CrossRef] [PubMed]
  56. Beauperin, M.; Top, S.; Richard, M.A.; Plażuk, D.; Pigeon, P.; Toma, S.; Poláčková, V.; Jaouen, G. The Length of the Bridging Chain in Ansa-Metallocenes Influences Their Antiproliferative Activity Against Triple Negative Breast Cancer Cells (TNBC). Dalton Trans. 2016, 45, 13126–13134. [Google Scholar] [CrossRef] [PubMed]
  57. Biancalana, L.; Zacchini, S.; Ferri, N.; Lupo, M.G.; Pampaloni, G.; Marchetti, F. Tuning the Cytotoxicity of Ruthenium(Ii) Para-Cymene Complexes by Mono-Substitution at a Triphenylphosphine/Phenoxydiphenylphosphine Ligand. Dalton Trans. 2017, 46, 16589–16604. [Google Scholar] [CrossRef] [PubMed]
  58. Becceneri, A.B.; Popolin, C.P.; Plutin, A.M.; Maistro, E.L.; Castellano, E.E.; Batista, A.A.; Cominetti, M.R. The Trans-[Ru(PPh3)2(N,N-dimethyl-N’-thiophenylthioureato-k2O,S)(bipy)]PF6 Complex Has Pro-Apoptotic Effects on Triple Negative Breast Cancer Cells and Presents Low Toxicity in Vivo. J. Inorg Biochem. 2018, 186, 70–84. [Google Scholar] [CrossRef] [PubMed]
  59. Popolin, C.P.; Reis, J.P.B.; Becceneri, A.B.; Graminha, A.E.; Almeida, M.A.P.; Corrêa, R.S.; Colina-Vegas, L.A.; Ellena, J.; Batista, A.A.; Cominetti, M.R. Cytotoxicity and Anti-Tumor Effects of New Ruthenium Complexes on Triple Negative Breast Cancer Cells. PLoS ONE 2017, 12, e0183275. [Google Scholar] [CrossRef] [PubMed]
  60. Chen, H.; Burnett, J.; Zhang, F.; Zhang, J.; Paholak, H.; Sun, D. Highly Crystallized Iron Oxide Nanoparticles as Effective and Biodegradable Mediators for Photothermal Cancer Therapy. J. Mater. Chem. B 2014, 2, 757–765. [Google Scholar] [CrossRef]
  61. Paholak, H.J.; Stevers, N.O.; Chen, H.; Burnett, J.P.; He, M.; Korkaya, H.; McDermott, S.P.; Deol, Y.; Clouthier, S.G.; Luther, T.; et al. Elimination of Epithelial-Like and Mesenchymal-Like Breast Cancer Stem Cells to Inhibit Metastasis Following Nanoparticle-Mediated Photothermal Therapy. Biomaterials 2016, 104, 145–157. [Google Scholar] [CrossRef] [PubMed]
  62. Manigandan, A.; Handi, V.; Sundaramoorthy, N.S.; Dhandapani, R.; Radhakrishnan, J.; Sethuraman, S.; Subramanian, A. Responsive Nanomicellar Theranostic Cages for Metastatic Breast Cancer. Bioconjug. Chem. 2018, 29, 275–286. [Google Scholar] [CrossRef] [PubMed]
  63. Xie, W.; Gao, Q.; Guo, Z.; Wang, D.; Gao, F.; Wang, X.; Wei, Y.; Zhao, L. Injectable and Self-Healing Thermosensitive Magnetic Hydrogel for Asynchronous Control Release of Doxorubicin and Docetaxel to Treat Triple-Negative Breast Cancer. ACS Appl. Mater. Interfaces 2017, 9, 33660–33673. [Google Scholar] [CrossRef] [PubMed]
  64. Harmon, T.; Harbuzariu, A.; Lanier, V.; Lipsey, C.C.; Kirlin, W.; Yang, L.; Gonzalez-Perez, R.R. Nanoparticle-Linked Antagonist for Leptin Signaling Inhibition in Breast Cancer. World J. Clin. Oncol. 2017, 8, 54–66. [Google Scholar] [CrossRef] [PubMed]
  65. Vyas, D.; Lopez-Hisijos, N.; Gandhi, S.; El-Dakdouki, M.; Basson, M.D.; Walsh, M.F.; Huang, X.; Vyas, A.K.; Chaturvedi, L.S. Doxorubicin-Hyaluronan Conjugated Super-Paramagnetic Iron Oxide Nanoparticles (DOX-HA-SPION) Enhanced Cytoplasmic Uptake of Doxorubicin and Modulated Apoptosis, IL-6 Release and NF-KappaB Activity in Human MDA-MB-231 Breast Cancer Cells. J. Nanosci. Nanotechnol. 2015, 15, 6413–6422. [Google Scholar] [CrossRef] [PubMed]
  66. Swanner, J.; Mims, J.; Carroll, D.L.; Akman, S.A.; Furdui, C.M.; Torti, S.V.; Singh, R.N. Differential Cytotoxic and Radiosensitizing Effects of Silver Nanoparticles on Triple-Negative Breast Cancer and Non-Triple-Negative Breast Cells. Int. J. Nanomed. 2015, 10, 3937–3953. [Google Scholar] [CrossRef]
  67. Juarez-Moreno, K.; Gonzalez, E.B.; Girón-Vazquez, N.; Chávez-Santoscoy, R.A.; Mota-Morales, J.D.; Perez-Mozqueda, L.L.; Garcia-Garcia, M.R.; Pestryakov, A.; Bogdanchikova, N. Comparison of Cytotoxicity and Genotoxicity Effects of Silver Nanoparticles on Human Cervix and Breast Cancer Cell Lines. Hum. Exp. Toxicol. 2017, 36, 931–948. [Google Scholar] [CrossRef] [PubMed]
  68. Hearn, J.M.; Hughes, G.M.; Romero-Canelón, I.; Munro, A.F.; Rubio-Ruiz, B.; Liu, Z.; Carragher, N.O.; Sadler, P.J. Pharmaco-Genomic Investigations of Organo-Iridium Anticancer Complexes Reveal Novel Mechanism of Action. Metallomics 2018, 10, 93–107. [Google Scholar] [CrossRef] [PubMed]
  69. Yang, G.J.; Zhong, H.J.; Ko, C.N.; Wong, S.Y.; Vellaisamy, K.; Ye, M.; Ma, D.L.; Leung, C.H. Identification of a Rhodium(III) Complex as a Wee1 Inhibitor Against TP53-Mutated Triple-Negative Breast Cancer Cells. Chem. Commun. 2018, 54, 2463–2466. [Google Scholar] [CrossRef] [PubMed]
  70. Mooney, R.; Roma, L.; Zhao, D.; van Haute, D.; Garcia, E.; Kim, S.U.; Annala, A.J.; Aboody, K.S.; Berlin, J.M. Neural Stem Cell-Mediated Intratumoral Delivery of Gold Nanorods Improves Photothermal Therapy. ACS Nano 2014, 8, 12450–12460. [Google Scholar] [CrossRef] [PubMed]
  71. Lacroix, M.; Leclercq, G. Relevance of Breast Cancer Cell Lines as Models for Breast Tumours: An Update. Breast Cancer Res. Treat. 2004, 83, 249–289. [Google Scholar] [CrossRef] [PubMed]
  72. Holliday, D.L.; Speirs, V. Choosing the Right Cell Line for Breast Cancer Research. Breast Cancer Res. 2011, 13, 215. [Google Scholar] [CrossRef] [PubMed]
  73. Rock, K.L.; Kono, H. The Inflammatory Response to Cell Death. Annu. Rev. Pathol. 2008, 3, 99–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Mammucari, C.; Rizzuto, R. Signaling Pathways in Mitochondrial Dysfunction and Aging. Mech. Ageing Dev. 2010, 131, 536–543. [Google Scholar] [CrossRef] [PubMed]
  75. Sohur, U.S.; Dixit, M.N.; Chen, C.L.; Byrom, M.W.; Kerr, L.A. Rel/NF-KappaB Represses Bcl-2 Transcription in Pro-B Lymphocytes. Gene Expr. 1999, 8, 219–229. [Google Scholar] [PubMed]
  76. Selvakannan, P.R.; Mandal, S.; Pasricha, R.; Adyanthaya, S.D.; Sastry, M. One-Step Synthesis of Hydrophobized Gold Nanoparticles of Controllable Size by the Reduction of Aqueous Chloroaurate Ions by Hexadecylaniline at the Liquid-Liquid Interface. Chem. Commun. 2002, 13, 1334–1335. [Google Scholar] [CrossRef]
  77. Okitsu, K.; Yue, A.; Tanabe, S.; Matsumoto, H.; Yobiko, Y. Formation of Colloidal Gold Nanoparticles in an Ultrasonic Field: Control of Rate of Gold (III) Reduction and Size of Formed Gold Particles. Langmuir 2001, 17, 7717–7720. [Google Scholar] [CrossRef]
  78. Korbekandi, H.; Ashari, Z.; Iravani, S.; Abbasi, S. Optimization of Biological Synthesis of Silver Nanoparticles Using Fusarium Oxysporum. Iran J. Pharm. Res. 2013, 12, 289–298. [Google Scholar] [PubMed]
  79. Iravani, S.; Korbekandi, H.; Mirmohammadi, S.V.; Zolfaghari, B. Synthesis of Silver Nanoparticles: Chemical, Physical and Biological Methods. Res. Pharm. Sci. 2014, 9, 385–406. [Google Scholar] [PubMed]
  80. Lesniak, A.; Kilinc, D.; Rashdan, S.A.; von Kriegsheim, A.; Ashall, B.; Zerulla, D.; Kolch, W.; Lee, G.U. In Vitro Study of the Interaction of Heregulin-Functionalized Magnetic-Optical Nanorods with MCF7 and MDA-MB-231 Cells. Faraday Discuss. 2014, 175, 189–201. [Google Scholar] [CrossRef] [PubMed]
  81. Greish, K. Enhanced Permeability and Retention (EPR) Effect for Anticancer Nanomedicine Drug Targeting. Methods Mol. Biol. 2010, 624, 25–37. [Google Scholar] [CrossRef] [PubMed]
  82. Matsumura, Y.; Maeda, H. A New Concept for Macromolecular Therapeutics in Cancer Hemotherapy: Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs. Cancer Res. 1986, 46, 6387–6392. [Google Scholar] [PubMed]
  83. Maeda, H.; Wu, J.; Sawa, T.; Matsumura, Y.; Hori, K. Tumor Vascular Permeability and the EPR Effect in Macromolecular Therapeutics: A Review. J. Control. Release 2000, 65, 271–284. [Google Scholar] [CrossRef]
  84. Dam, D.H.; Culver, K.S.; Kandela, I.; Lee, R.C.; Chandra, K.; Lee, H.; Mantis, C.; Ugolkov, A.; Mazar, A.P.; Odom, T.W. Biodistribution and in Vivo Toxicity of Aptamer-Loaded Gold Nanostars. Nanomedicine 2015, 11, 671–679. [Google Scholar] [CrossRef] [PubMed]
  85. Cioce, M.; Gherardi, S.; Viglietto, G.; Strano, S.; Blandino, G.; Muti, P.; Ciliberto, G. Mammosphere-Forming Cells from Breast Cancer Cell Lines as a Tool for the Identification of CSC-Like- and Early Progenitor-Targeting Drugs. Cell Cycle 2010, 9, 2878–2887. [Google Scholar] [CrossRef] [PubMed]
  86. Rosen, J.M.; Jordan, C.T. The Increasing Complexity of the Cancer Stem Cell Paradigm. Science 2009, 324, 1670–1673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bravo-Cordero, J.J.; Hodgson, L.; Condeelis, J. Directed Cell Invasion and Migration during Metastasis. Curr. Opin. Cell Biol. 2012, 24, 277–283. [Google Scholar] [CrossRef] [PubMed]
  88. Anampa, J.; Sparano, J.A. New Agents for the Management of Resistant Metastatic Breast Cancer. Expert Opin. Pharmacother. 2017, 18, 1815–1831. [Google Scholar] [CrossRef] [PubMed]
  89. Toraya-Brown, S.; Sheen, M.R.; Zhang, P.; Chen, L.; Baird, J.R.; Demidenko, E.; Turk, M.J.; Hoopes, P.J.; Conejo-Garcia, J.R.; Fiering, S. Local Hyperthermia Treatment of Tumors Induces CD8(+) T Cell-Mediated Resistance Against Distal and Secondary Tumors. Nanomedicine 2014, 10, 1273–1285. [Google Scholar] [CrossRef] [PubMed]
  90. Aggarwal, B.B.; Shishodia, S.; Sandur, S.K.; Pandey, M.K.; Sethi, G. Inflammation and Cancer: How Hot Is the Link? Biochem. Pharmacol. 2006, 72, 1605–1621. [Google Scholar] [CrossRef] [PubMed]
  91. Salgado, R.; Junius, S.; Benoy, I.; van Dam, P.; Vermeulen, P.; van Marck, E.; Huget, P.; Dirix, L.Y. Circulating Interleukin-6 Predicts Survival in Patients with Metastatic Breast Cancer. Int. J. Cancer 2003, 103, 642–646. [Google Scholar] [CrossRef] [PubMed]
  92. Berberoglu, U.; Yildirim, E.; Celen, O. Serum Levels of Tumor Necrosis Factor Alpha Correlate with Response to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer. Int. J. Biol. Markers 2004, 19, 130–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hartmann, L.C.; Keeney, G.L.; Lingle, W.L.; Christianson, T.J.; Varghese, B.; Hillman, D.; Oberg, A.L.; Low, P.S. Folate Receptor Overexpression Is Associated with Poor Outcome in Breast Cancer. Int. J. Cancer 2007, 121, 938–942. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the metastatic process in triple negative breast cancer (TNBC) and its inhibition by non-platinum metal-based complexes (NPMBC). Metastasis is a sequential and interrelated multi-step process that consists of four main essential steps: migration to adjacent tissue after detachment from a primary tumor; local invasion of cancer cells into adjacent tissue; trans-endothelial migration of cancer cells into vessels (intravasation); transport of cancer cells through the circulatory system with extravasation of them in a secondary organ (brain, lung, liver, bone). The anti-metastatic effect of NPMBC has been associated with their ability: to inhibit matrix metalloproteinases (a); to inhibit mesenchymal markers such as vimentin (b); to interfere with the function of integrins (c); and to modify the immune/inflammatory response (d,e).
Figure 1. Schematic representation of the metastatic process in triple negative breast cancer (TNBC) and its inhibition by non-platinum metal-based complexes (NPMBC). Metastasis is a sequential and interrelated multi-step process that consists of four main essential steps: migration to adjacent tissue after detachment from a primary tumor; local invasion of cancer cells into adjacent tissue; trans-endothelial migration of cancer cells into vessels (intravasation); transport of cancer cells through the circulatory system with extravasation of them in a secondary organ (brain, lung, liver, bone). The anti-metastatic effect of NPMBC has been associated with their ability: to inhibit matrix metalloproteinases (a); to inhibit mesenchymal markers such as vimentin (b); to interfere with the function of integrins (c); and to modify the immune/inflammatory response (d,e).
Crystals 08 00369 g001
Table 1. Characterization of the main triple negative breast cancer (TNBC) cell lines according to the subtype and molecular features.
Table 1. Characterization of the main triple negative breast cancer (TNBC) cell lines according to the subtype and molecular features.
SubtypeTNBC Cell LinesOthers Immunohistochemical Markers besides ER, PR and HER2 aTumor of Origin
ABT-20WNT3 oncogene +, WNT7B oncogene +Invasive ductal carcinoma
CAL148CK 7 +, CK 8 +, CK 17 − CK 18 +, CK 19 +, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin −Adenocarcinoma
DU4475CK 7 −, CK 8 +, CK 17 −, CK 18 +, CK 19 −, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin −Invasive ductal carcinoma
EMG3CK 5 +, CK 14 +, CK 18 +, CK 19 −, EMA +, P63+, SMA −, ESA +, EGFR +Invasive ductal carcinoma
HCC1143p53 +, EGP2 +, CK 19 +Ductal carcinoma
HCC1187EGP2 +, CK 19 +, p53 +++Ductal carcinoma
HCC1599EGP2 +, CK 19 +, p53 −Ductal carcinoma
HCC1806p53 −, EGP2 +, CK 19 +, Ob-R +Squamous carcinoma
HCC1937p53 −, EGP2 +, CK 19 +Ductal carcinoma
HCC2157EGP2 +, CK 19 +, p53 +Ductal carcinoma
HCC3153N.A.Ductal carcinoma
HCC70p53 +++, EGP2 + CK 19 +Ductal carcinoma
HMT3522Vimentin −, PLK1 +Benign tumor
KPL-3CKeratins −, Vimentin −Invasive ductal carcinoma
MA11e-cadherin +Invasive lobular carcinoma
MDA-MB-436Tubulin +, actin +, ICAM +++Adenocarcinoma
MDA-MB-468EGFR +, TGF alpha +, Ki67 highAdenocarcinoma
MFM223CK 7 +, CK 8 +, CK 17 −, CK 18 +, CK 19 +, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin −Carcinoma
SUM229PECK 8 +, CK 18 +, CK 19 +Ductal carcinoma
BA17Vimentin +, CK 14 +, N-cadherin +, COX2 +++Carcinoma
BCM-2665AALDH +Invasive ductal carcinoma
BT-549Ki67 low, e-cadherin low, claudin-3 low, claudinin-4 low, claudinin-7 lowInvasive ductal carcinoma
CAL-120CK 7 −, CK 8 +, CK 17 −, CK 18 +, CK 19 +, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin +Adenocarcinoma
CAL-51CK 7 −, CK 8 +, CK 17 −, CK 18 +, CK 19 +, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin +Adenocarcinoma
CAL-85-1CK 7 +, CK 8 +, CK 17 +, CK 18 +, CK 19 +, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin +Adenocarcinoma
HCC1395p53 +, EGP2 +, CK 19 +Ductal carcinoma
HCC1739N.A.Ductal carcinoma
HCC38p53 +, EGP2 +, CK 19 +Ductal carcinoma
HDQ-P1CK 7 +, CK 8 +, CK 17 +, CK 18 +, CK 19 +, desmin −, endothel −, EpCAM +, GFAP −, neurofilament −, vimentin +Invasive ductal carcinoma
Hs578TKi67 low, e-cadherin low, claudin-3 low, claudinin-4 low, claudinin-7 low, EGFR +Invasive ductal carcinoma
MDA-MB-157WNT7B oncogene +, ICAM +++Medullary carcinoma
MDA-MB-231EGFR +, TGF alpha +, CD44 +++, LHRH +++, WNT7B oncogene +, ICAM-1 +++, Ki67 low, e-cadherin low, claudin-3 low, claudinin-4 low, claudinin-7 low, alpha-V beta-3 integrin +, Ob-R +Adenocarcinoma
OCUB-Me-cadherin +, Laminin +Carcinoma
SUM1315EGFR +Carcinoma
SUM149CK 8 +, CK 18 +, CK 19 +Carcinoma
SUM159claudin low, CK 8 +, CK 18 +, CK 19 +Carcinoma
The correspondence between the cell lines and clinical features of the tumor of origin is provided. a Estrogen receptors (ER), progesterone receptors (PR) and human epidermal growth factor receptor 2 (HER2) are all negative. Abbreviations: −, Negative; +, Positive; +++, Overexpression; ALDH, Aldehyde Dehydrogenase; CK, Cytokeratin; COX, Cyclooxygenase; EGF, Epidermal Growth Factor; EGFR, Epidermal Growth Factor Receptor; EGP, Epithelial Glycoprotein; EMA, Epithelial Membrane Antigen; EpCAM, Epithelial Cell Adhesion Molecule; ESA, Epithelial Specific Antigen; GFAP, Glial Fibrillary Acidic Protein; ICAM, Intercellular Adhesion Molecule; LHRH, Luteinizing Hormone-Releasing Hormone; N.A., Not Available; Ob-R, Leptin Receptor; PLK, Polo-Like Kinase; SMA, Smooth Muscle Actin; TGF alpha, Transforming growth factor alpha [4,10,11,12,13].
Table 2. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inhibiting tumor cell proliferation.
Table 2. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inhibiting tumor cell proliferation.
In Vitro
Cell LineNPMBCAssayMode of Action/Pathway InvolvedEffect ObservedDoses AssayedReference
MDA-MB-231Multibranched gold nanoantennasCalcein/PI staining assay---Induction of cell death170 μg/mL[14]
MDA-MB-231
SUM159
MDA-MB-468
BT-549
Liposome encapsulated ruthenium polypyridine complexMTT assay---Reduced cell viability <4 µM [16]
MDA-MB-231Liposome encapsulated ruthenium polypyridine complexWestern blot
Immunofluorescence
γ-H2AX
ATM
Increased levels of γ-H2AX
Degradation and dephosphorylation of ATM
3 µM[16]
MDA-MB-231Gold nanostars loaded with AS1411Resazurin cell viability assay---40% cell viability0.3 nM[27]
MDA-MB-231CD44-targeting HSP72 depletion nanosystem based on plasmonic gold nanoparticlesMTT assay---20–85% cell death50 mg/L[28]
MDA-MB-231Multibranched gold nanoantennas MTT assay---~40% cell viability 170 μg/mL[29]
MDA-MB-231Gold nanoparticlesMTT assay---15–75% survival0.1–100 µM[31]
MDA-MB-231Thioglucose-bound gold nanoparticlesMTT assay---Inhibited cell growth20 nM[36]
MDA-MB-231Thioglucose-bound gold nanoparticlesClonogenic cell survival assay---Enhanced radiosensitivity 20 nM[36]
MDA-MB-231
MDA-MB-468
Anti-EGFR-gold nanorod conjugatesMTT assay---Inhibited cell growth 1.84 µg/mL[37]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesWestern blotKi-67
EGFR
Akt
ERK1/2
mTOR
FAK
Decreased Ki-67 and EGFR expression Suppression of phosphorylation of FAK, ERK1/2, mTOR and Akt1.84 µg/mL[37]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesCCK-8 assay---32.2% cell viability 120 pM[38]
MDA-MB-231Gold nanospheres conjugated with AS1411MTT assay---Inhibited cell viability <100 nM[39]
MDA-MB-231
SUM1315
HCC1937
MDA-MB-468
Gold-nanoparticles conjugated with Rad6 inhibitorMTT assay
Trypan blue staining assay
---Decreased cell viability (SUM1315 and MDA-MB-231)
No change in viability of HCC1937 and MDA-MB-468
0.5 µM for SUM1315; 8.2 µM for MDA-MB-231 [40]
MDA-MB-231Gold nanoparticles loaded with 5-fluorouracilMTT assay---Decreased cell viability0.5 nM [41]
MDA-MB-231Chitosan layered gold nanorodsMTS-based colorimetric assay---~65% inhibition of cell proliferation100 nM[43]
MDA-MB-231
MDA-MB-468
Micellar gold nanoparticles conjugated with ZD6474MTT assay---80.51% (MDA-MB-231) and ~92% (MDA-MB-468) inhibition of cell proliferation1–10 µM[44]
MDA-MB-231
BT-20
Copper salicylate phenanthroline complexesMTS-based colorimetric assay---Inhibition of cell growth5–25 µM[49]
MDA-MB-231Copper complexes of phenanthrenequinone thiosemicarbazoneMTT assay---Decreased cell viability2.3–4.8 µM [50]
MDA-MB-231Nickel complexes of phenanthrenequinone thiosemicarbazoneMTT assay---Decreased cell viability>10 µM[50]
MDA-MB-231Palladium complexes of phenanthrenequinone thiosemicarbazoneMTT assay---Decreased cell viability>10 µM[50]
MDA-MB-231Copper oxide nanowire conjugated with folic acidFlow cytometry---60% cell death2.5 µg/mL[51]
MDA-MB-231Water-soluble iminophosphorane ruthenium(II) compoundsMTT assay---Decreased cell viability2.61–75.4 µM [52]
MDA-MB-231
MDA-MB-468
MDA-MB-436
Ruthenium-based nucleolipidic nanoaggregatesMTT assay
Trypan blue
---Reduced cell proliferation 12.1–14.7 µM [53]
MDA-MB-231
HCC1937
Ruthenium complexesReal-time cell growth profiling
MTT assay
---Inhibited cell viability 13.2 and 14.1 µM for MDA-MB-231; 1.8 and 9.9 µM for HCC1937 [54]
A17
MDA-MB-231
Ruthenium complexesMTT assay---Inhibited cell viability 230.66 µM for A17; 409.89 µM for MDA-MB-231 [55]
MDA-MB-231Ansa-ferrocenes ruthenocenophanesMTT assay---Decreased cell viability0.09–4.53 µM [56]
MDA-MB-231Ruthenium(II) paracyme complexesSulforhodamine B assay---Decreased cell viability9.2–29.1 µM [57]
MDA-MB-231Ruthenium complexesMTT assay---Decreased cell viability8.81–21.92 µM [58]
MDA-MB-231Ruthenium complexesColony formation---100% inhibition of the colony formation2 µM[58]
MDA-MB-231Ruthenium-based compoundsMTT assay---Decreased cell viability31.16 and 52.74 µM [59]
MDA-MB-231Ruthenium-based compoundsColony formation---100% inhibition of the colony formation20 µM[59]
SUM159Iron oxide nanoparticlesAlamar blue cell viability assay
Flow cytometry
---5–80% cell viability 10 µM[61]
MDA-MB-231Supermagnetic iron oxide nanoparticles conjugated with doxorubicinMTS-based colorimetric assay---31%-cell viability520 µM[62]
MDA-MB-231Chitosan hydrogel cross-linked with telechelic difunctional poly(ethylene glycol) modified iron oxide magnetic nanoparticlesCCK-8 assay---5.6% cell viability10.8–35.8 µM [63]
MDA-MB-231
HCC1806
Iron oxide nanoparticles linked antagonist for leptinMTT assay---Prevention of leptin-induced cell proliferation0.0036 pmol/L[64]
MDA-MB-231
HCC1806
Iron oxide nanoparticles linked antagonist for leptinImmunoblotting analysisCyclin D1Prevention of leptin-induced cyclin D1 expression0.0036 pmol/L[64]
MDA-MB-231
HCC1806
Iron oxide nanoparticles linked antagonist for leptinCell cycle analysis---Prevention of leptin-induced cell cycle progression0.0018–0.0036 pmol/L for MDA-MB-231 and 0.0018–0.036 pmol/L for HCC1806[64]
MDA-MB-231
HCC 1806
Iron oxide nanoparticles linked antagonist for leptinFITC-annexin V/PI staining assay---Decreased cell viability 0.0036 pmol/L[64]
MDA-MB-231Doxorubicin-hyaluronan conjugated iron oxide nanoparticlesConfocal microscopyVimentin e-cadherinDecreased vimentin expression
No change in e-cadherin expression
2 µM[65]
MDA-MB-231
BT549
SUM-159
Silver nanoparticlesMTT assay---Inhibited cell viability≥10 µg/mL[66]
MDA-MB-231
BT549
SUM-159
Silver nanoparticlesClonogenic assay---100% inhibition of clonogenic growth≥10 µg/mL[66]
MDA-MB-231Silver nanoparticlesMTT assay---Decreased cell viability 6.72 µg/mL at 12 h and 2.62 µg/mL at 24 h [67]
MDA-MB-468
OCUB-M
Organo-iridium complexes
Organo-osmium complexes
MTT assay---Decreased cell viability<1 µM [68]
MDA-MB-468
MDA-MB-231
Rhodium(III) complexMTT assay---Inhibited cell proliferation0.35 and 0.55 µM[69]
MDA-MB-231Rhodium(III) complexWestern blotWee 1
Cdc2
ERK
Akt
Cyclin A2
γ-H2AX
Decreased phosphorylation of ERK, Akt, Cdc2
Decreased expression of cyclin A2
Increased γ-H2AX
levels
Inhibition of Wee1 activity
1–3 μM[69]
In Vivo
Cell LineNPMBCAnimal (Model)/Analysis from Animal TissueMode of Action/Pathway InvolvedEffect ObservedDoses Assayed (Route of Administration)Reference
MDA-MB-231Liposome encapsulated ruthenium polypyridine complexAthymic nude mice (orthotopic)/Immunofluorescen imagingKi-67Decreased Ki-67 level5 mg ruthenium/kg (i.v.)[16]
MDA-MB-231Liposome encapsulated ruthenium polypyridine complexAthymic nude mice (orthotopic)---Suppression of tumor growth (tumor weights: 0.342 with nanoparticles vs. 0.992 with control) 5 mg ruthenium/kg (i.v.)[16]
MDA-MB-231CD44-targeting HSP72 depletion nanosystem based on plasmonic gold nanoparticlesNude mice (xenograft)---Smaller tumor volume than control10 mg/kg (i.v.)[28]
MDA-MB-231Gold nanomatryoshkas and nanoshellsSprague-Dawley athymic nude mice (xenograft)---Improved survival (15.8 and 11.3 days with nanomatryoshkas and nanoshells respectively)
Slower tumor growth with nanomatryoshkas
300 µg of gold (i.v.)[32]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesBALB/c nude mice (xenograft)---Smaller tumor volumen than control (~<50 vs. 250 mm3 respectively)0.5 pmol/g mouse (i.v.)[37]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesBALB/c nude mice (xenograft)/ImmunohistochemistryKi-67
EGFR
Decreased Ki-67 and EGFR levels0.5 pmol/g mouse (i.v.)[37]
MDA-MB-231Gold nanospheres conjugated with AS1411Nude mice Fox1nu (xenograft)---Decreased tumor grow rate and tumor regression (tumor size at 12 days: 100 with nanospheres vs. 1000 mm3 with control)1 mg/kg/day (i.p.)[39]
MDA-MB-231Hydrogel embedded with gold nanoparticlesSCID hairless congenic mice (orthotopic)---Efficient and sustained inhibition of tumor growth (~90% tumor size reduction)10 nM of gold nanoparticles (i.h.)[41]
MDA-MB-231Gold nanomatryoshkas and nanoshellsSprague-Dawley athymic nude mice (xenograft)---Improved survival (83% and 33% at 60 days with nanomatryoshkas and nanoshells respectively)
Complete tumor regression at 60 days post-treatment with relapse rates between 16.7% (nanomatryoshkas) and 50% (nanoshells)
300 µg of gold (i.v.)[42]
MDA-MB-231Micellar gold nanoparticles conjugated with ZD6474Athymic BALB/c (nu+/nu+) mice (xenograft)---Decrease in tumor volume (63% of control)30 mg/kg of ZDD6474 (i.v.)[44]
BCM-2665AGold nanoshellsSCID/beige mice (xenograft)---Smaller tumor volume compared to control (400 vs. 800 mm3 respectively)~8 × 108 nanoshells/g body weight (i.v.)[45]
MDA-MB-231RGD-conjugated mesoporous silica-encapsulated gold nanorodsNude mice (orthotopic)---Smaller tumor volume than control (569 vs. 1302 mm3 respectively)∼50 μg of gold/g of body weight (i.v.)[46]
MDA-MB-231Copper salicylate phenanthroline complexesNude mice (xenograft)---Smaller tumor volume than control (20 vs. 35 mm3 respectively)0.5 mg/mL (i.p.)[49]
MDA-MB-231Copper salicylate phenanthroline complexesNude mice (xenograft)/Immunohistochemical stainingKi-67Reduced Ki-67 expression0.5 mg/mL (i.p.)[49]
MDA-MB-231Water-soluble iminophosphorane ruthenium(II) compoundsNOD.CB17-Prkdc SCID/J mice (xenograft)---Significant inhibition of tumor growth and smaller tumor size than control (56% decrease vs. 200% increase in tumor volume respectively)5 mg/kg/day (i.p.)[52]
A17Ruthenium complexesFVB/NCrl mice (xenograft)---Decreased tumor grow rate and smaller tumor size than control (<3 vs. 7 mm) 210 mg/kg/day (i.p.)[55]
SUM159Highly crystallized iron oxide nanoparticlesBALB/c immune-competent mice (xenograft)---Complete tumor regression without relapses20 mg iron/kg body weight (i.v.)[60]
SUM159Iron oxide nanoparticlesNOD/SCID immune-compromised mice (orthotopic)---Smaller tumor size than control20 mg iron/kg body weight (i.v.)[61]
4T1Supermagnetic iron oxide nanoparticles conjugated with doxorubicinBALB/c mice (orthotopic)---Smaller tumor size than control (7 vs. 74 mm3 respectively)0.25 mg iron/100 mm3 (i.t.)[62]
MDA-MB-231Chitosan hydrogel cross-linked with telechelic difunctional poly(ethylene glycol) modified iron oxide magnetic nanoparticlesBALB/c mice (xenograft)---Smaller tumor size than control (relative tumor volume ~0.75 vs. 4.5 v/v respectively)18.7 mg/kg (i.t.)[63]
MDA-MB-231Silver nanoparticlesnu/nu athymic mice (xenograft)---Decreased tumor grow rate and smaller tumor size than control (250 vs. 550 mm3 respectively)0.2 µg/mm3 tumor volume (i.t.)[66]
MDA-MB-231Gold nanorod-loaded neural stem cellsAthymic nude mice (xenograft)---Complete tumor ablation, improved survival and reduced tumor recurrence compared to control12.5 µg/(i.t.)[70]
Abbreviations: ---, Not analyzed; Akt, Protein Kinase B; ATM, Ataxia Telangiectasia Mutated Protein; CCK-8, Cell Counting Kit-8; Cdc2, Cell Division Control Protein 2 Homolog; EGFR, Epidermal Growth Factor Receptor; ERK, Extracellular Signal–Regulated Kinase; FAK, Focal Adhesion Kinase; Fox 1, Forkhead Box Protein 1; HSP72, Heat Shock Protein 72; i.h., implantation of a hydrogel disk on top of the tumors; i.p., intraperitoneal injection; i.t., intratumoral injection; i.v., intravenous injection; MRP1, Multidrug Resistance Protein 1; mTOR, Mammalian Target of Rapamycin; MTS, 3-(4,5-Dimethylthiazol-2-yl)-5-(3-Carboxymethoxyphenyl)-2(4-Sulfonyl)-2H-Tetrazolium; MTT, 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide; NOD, Non-Obese Diabetic; PI, Propidium Iodide; SCID, Severe Combined Immunodeficient.
Table 3. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inducing apoptosis.
Table 3. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inducing apoptosis.
In Vitro
Cell LineNPMBCAssayMode of Action/Pathway InvolvedEffect ObservedDoses AssayedReference
MDA-MB-231Liposome encapsulated ruthenium polypyridine complexWestern blot
PARP
Caspase 3
Caspase 8
Caspase 9
Increased cleaved PARP, caspase 3, caspase 8 and caspase 9 levels5 µM[16]
MDA-MB-231Liposome encapsulated ruthenium polypyridine complexFlow cytometry (cell cycle arrest)---75.9% apoptotic cells
54% cells in the G2/M phase
5 µM[16]
MDA-MB-231Gold nanostarsFluorescent assay
qPCR
Caspase 3/7
Bcl-2
Increased caspase activity
Downregulation of Bcl-2 expression
0.3 nM[27]
MDA-MB-231Multibranched gold nanoantennasAnnexin V/calcein/PI staining assay---Induction of apoptotic cell death 170 μg/mL[29]
MDA-MB-231Gold nanoparticles conjugated gemcitabineAnnexin V/7-AAD assay---Induction of apoptosis0.5, 1, 5, 25 nM[30]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesFITC-annexin V/PI staining assay---~25–40% apoptotic cells1.84 µg/mL[37]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesWestern blotCaspase 3Increased cleaved caspase 3 protein1.84 µg/mL[37]
MDA-MB-231Gold nanospheresFITC-annexin V/PI staining assay---88% apoptotic cells200 nM[39]
MDA-MB-231
SUM1315
HCC1937
MDA-MB-468
Gold-nanoparticles conjugated with Rad6 inhibitorAcridine orange/ethidium bromide staining---SUM1315 and MDA-MB-231 cells in early and late stages of apoptosis
No morphological alterations consistent with apoptosis were observed in HCC1937 or MDA-MB-468 cells
1–5 μM [40]
SUM1315
HCC1937
Gold-nanoparticles conjugated with Rad6 inhibitorMitochondrial membrane potential (JC-1)---Loss in the mitochondrial function
of SUM1315 cells
Mitochondrial function of HCC1937 cells was unaffected
1 µM[40]
SUM1315
HCC1937
Gold-nanoparticles conjugated with Rad6 inhibitorWestern blotPARP-1Activation of PARP-11–5 μM[40]
MDA-MB-468Micellar gold nanoparticles conjugated with ZD6474FITC-annexin V/PI staining assay---28.2% apoptotic cells5 µM[44]
MDA-MB-231RGD-conjugated mesoporous silica-encapsulated gold nanorodsFITC-annexin V/PI staining assay---Increased apoptotic cells50 µg/mL[46]
MDA-MB-231RGD-conjugated mesoporous silica-encapsulated gold nanorodsDHE fluorescence methodROSIncreased cellular ROS levels50 µg/mL[46]
MDA-MB-231
BT-20
Copper salicylate phenanthroline complexesFlow cytometry (Annexin V/PI)--->80% apoptotic cells25 µM[49]
MDA-MB-231
BT-20
Copper salicylate phenanthroline complexesWestern blotBcl-2
Bcl-xL
Survivin
PARP
Decreased Bcl-2, Bcl-xL and surviving expression
Increased cleaved PARP expression
25 μM[49]
MDA-MB-231Copper oxide nanowire conjugated with folic acidFITC-annexin V/PI staining assay---Induction of apoptosis2.5 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidSEM---Formation of apoptotic bodies and membrane blebbing2.5 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidFlow cytometry---Induction of ROS2.5 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidMitochondrial membrane potential (JC-1)
Immunofluorescence
Western blotting analysis
ROS
Cyt C
Loss in the mitochondrial membrane potential
Increased expression of Cyt C in the cytosol
2.5 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidWestern blotting analysisCaspase 3
Caspase 9
Increased cleaved caspase 9 and caspase 3 expression2.5 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidWestern blotting analysis
Immuno-fluorescence staining
NF-κB p65 proteinInhibition of nuclear transport of the p65 subunit of NF-κB protein2.5 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidqRT-PCR
Western blot
miR425
PTEN
Akt
Down-regulation of miR-425 and Akt, and up-regulation of PTEN2.5 µg/mL[51]
MDA-MB-231Ruthenium-based nucleolipidic nanoaggregatesPhase-contrast microscopy---Cell shrinkage and loss of cell-cell contact14.7 µM[53]
MDA-MB-231Ruthenium-based nucleolipidic nanoaggregatesFITC-annexin V/PI staining assay---~80% of cells in late apoptosis phase14.7 µM [53]
MDA-MB-231Ruthenium-based nucleolipidic nanoaggregatesDNA fragmentation assay---Internucleosomal DNA laddering14.7 µM[53]
MDA-MB-231Ruthenium-based nucleolipidic nanoaggregatesWestern blotBax
Bcl-2
Caspase 3
Caspase 9
Caspase 8
Up-regulated Bax expression
Down-regulated Bcl-2 expression
Activation of caspase 9, caspase 8 and caspase 3
14.7 µM[53]
MDA-MB-231
HCC1937
Ruthenium complexesRT-PCRP21
p53
BRCA1
Upregulated p53, P21 and BRCA1 expression13.2 and 14.1 µM for MDA-MB-231;
1.8 and 9.9 µM for HCC1937
[54]
A17
MDA-MB-231
Ruthenium complexesImmunohistochemistry
Western blot
Caspase 3Increased number of apoptotic cells (cleaved caspase 3 positive cells)750 μM for A17; 1 mM for MDA-MB-231[55]
MDA-MB-231Ruthenium complexesDAPI staining---Nuclear chromatin condensation with formation of apoptotic bodies 2, 4 and 8 µM[58]
MDA-MB-231Ruthenium complexesFlow cytometry (PE annexin-V)---43% apoptotic cells2 µM[58]
MDA-MB-231Ruthenium complexesqRT-PCRBax
Caspase 3
Bcl-2
Increased Bax and caspase 3 expression
Decreased Bcl-2 expression
4, 8 and 16 µM[58]
MDA-MB-231Ruthenium complexesWestern blottingBax
Caspase 3
Bcl-2
Increased Bax expression
Decreased Bcl-2 expression
No change in caspase-3 expression
0.5, 1, 2, 4, 8 and 16 µM[58]
MDA-MB-231Ruthenium complexesComet assay---DNA damage4 and 8 µM[58]
MDA-MB-231Ruthenium-based compoundsDAPI staining---Nuclear fragmentation60 and 70 µM[59]
MDA-MB-231Ruthenium-based compoundsFlow cytometry (PE annexin-V)---74.4% apoptotic cells20 µM[59]
MDA-MB-231Ruthenium-based compoundsqRT-PCRBax
Caspase 3
Bcl-2
Increased expression of Bax and caspase 3.
No change in
gene expression of Bcl-2
40 µM[59]
MDA-MB-231Ruthenium-based compoundsWestern blottingCaspases 3 and 9Increased caspase 3 and 9 expression 2.5 µM[59]
MDA-MB-231Ruthenium-based compoundsWestern blottingBcl-2No change in Bcl-2 expression10 µM[59]
MDA-MB-231Supermagnetic iron oxide nanoparticles conjugated with doxorubicinMitochondrial membrane potential (JC-1)---Increased depolarization of mitochondrial membrane potential520 µM[62]
MDA-MB-231
HCC 1806
Iron oxide nanoparticles linked antagonist for leptinImmunoblotting analysisSTAT3Prevention of leptin-induced STAT3 expression only in HCC1806 cells0.0036 pmol/L[64]
MDA-MB-231Doxorubicin-hyaluronan conjugated iron oxide nanoparticlesFITC-annexin V/PI staining assay---12% apoptotic cells (early and late stages of apoptosis)2 µM[65]
MDA-MB-231Doxorubicin-hyaluronan conjugated iron oxide nanoparticlesSteady-Glo luciferase reagent assayNF-κBReduced NF-κB transcriptional activity (~40%)2 µM[65]
MDA-MB-231Silver nanoparticlesFITC-annexin V/PI staining assay---Cells in early and late stages of apoptosis1.25–10 µg/mL[67]
MDA-MB-231Silver nanoparticlesComet assay---DNA damage 5 and 10 µg/mL[67]
MDA-MB-231Silver nanoparticlesFlow cytometry
Confocal microscopy
Fluorescence microscopy
ROSIncrease ROS levels2.5, 5 and 10 µg/mL[67]
MDA-MB-231Rhodium(III) complexWestern blotCaspase 3
Caspase 7
Caspase 8
Bcl-2
PARP
Increased PARP, and cleaved caspases-3, -7, and -8 expression
Decreased Bcl-2 expression
1 and 3 µM[69]
In Vivo
Cell LineNPMBCAnimal (Model))/Analysis from Animal TissueMode of Action/Pathway InvolvedEffect ObservedDoses Assayed (Route of Administration)Reference
MDA-MB-231Liposome encapsulated ruthenium polypyridine complexAthymic nude mice (orthotopic))/Western blot, immunohistochemistry
PARP
Caspase 3
Caspase 8
Caspase 9
TUNEL
Increased TUNEL cleaved PARP, caspase 3, caspase 8 and caspase 9 levels 5 mg ruthenium/kg (i.v.)[16]
MDA-MB-231CD44-targeting HSP72 depletion nanosystem based on plasmonic gold nanoparticlesImmunohistochemistryTUNELMassive apoptotic cells10 mg/kg (i.v.)[28]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesBALB/c nude mice (xenograft))/
Immunohistochemistry
TUNEL
Caspase 3
Increased TUNEL and caspase 3 levels 0.5 pmol/g mouse (i.v.)[37]
MDA-MB-231Copper salicylate phenanthroline complexesNude mice (xenograft))/
Immunohistochemical staining
Bcl-2
Bcl-xL
Survivin
Reduced Bcl-2, survivin and Bcl-xL expression0.5 mg/mL (i.p.)[49]
4T1Copper oxide nanowire conjugated with folic acidImmunohistochemical analysis of tumors of BALB/c mice (orthotopic))---Suppression of tumor growth (lower tumor weight and volume)2.5 mg/kg body weight (i.p.)[51]
4T1Copper oxide nanowire conjugated with folic acidImmunohistochemical analysis of tumors of BALB/c mice (orthotopic))/Immunohistochemical analysis and western blotPTEN
miR425
Increased PTEN expression and decreased miR-425 expression2.5 mg/kg body weight (i.p.)[51]
A17Ruthenium complexesFVB/NCrl mice (xenograft))/Immunohistochemistry analysisCaspase 3Increased number of apoptotic cells in tumors (cleaved caspase-3 positive cells)210 mg/kg/day (i.p.)[55]
Abbreviations: ---, Not analyzed; 7-AAD, 7-Aminoactinomycin D; Akt, Protein Kinase B; Bax, Bcl-2-Like Protein 4; Bcl-2, B-Cell Lymphoma 2; Bcl-XL, B-Cell Lymphoma-Extra Large; BRCA1, Breast Cancer Susceptibility Gene 1; Cyt C, Cytochrome C; DAPI, 4′,6-Diamidino-2-Phenylindole; DHE, Dihydroethidium; EGFR, Epidermal Growth Factor Receptor; FITC, Fluorescein Isothiocyanate; HSP72, Heat Shock Protein 72; i.p., intraperitoneal injection; i.v., intravenous injection; miR425, microRNA425; NF-κB, Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells; p65 protein, Nuclear factor NF-κB P65 Subunit; PARP, Poly(ADP-ribose) POLYMERASE; PE, Phycoerythrin; PI, Propidium Iodide; PTEN, Phosphatase and Tensin Homolog; qRT-PCR, Quantitative Reverse Transcription Polymerase Chain Reaction; ROS, Reactive Oxygen Species; SEM, Scanning Electron Microscopy; STAT3, Signal Transducer and Activator of Transcription 3; TUNEL, Terminal Deosynucleotidyl Transferase dUTP Nick End Labeling.
Table 4. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inducing autophagic cell death.
Table 4. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inducing autophagic cell death.
In Vitro
Cell LineNPMBCAssayMode of Action/Pathway InvolvedEffect ObservedDoses AssayedReference
MDA-MB-231Anti-EGFR-gold nanorod conjugatesImmunofluorescence stainingLC3Increased LC3 expression120 pM[38]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesTEM---Induction of double- or multiple-membrane vesicles120 pM[38]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesWestern blotBeclin-1
Atg5
p62
LC3
Akt
mTOR
Increased Beclin-1, Atg5, LC3-II, and p62 expression
Decreased phosphorylation of mTOR and Akt
120 pM[38]
MDA-MB-231
SUM1315
Gold-nanoparticles conjugated with Rad6 inhibitorWestern blotLC3-I
LC3-II
p62
Increased conversion of LC3-I to LC3-II in SUM1315 cells
Decreased p62 expression in SUM1315 cells
No changes in LC3-I/II ratios or expression of p62 protein in MDA-MB-231 cells
1–5 μM[40]
MDA-MB-231Ruthenium-based nucleolipidic nanoaggregatesPhase-contrast microscopy
Monodansylcadaverine assay
---Increased formation of autophagic vacuoles12.1–14.7 µM [53]
In Vivo
Cell LineNPMBCAnimal (Model)/Analysis from Animal TissueMode of Action/Pathway InvolvedEffect ObservedDoses Assayed (Route of Administration)Reference
MDA-MB-231Anti-EGFR-gold nanorod conjugatesBALB/c nude mice (xenograft)---Significant decrease in tumor volume compared to control (120 vs. 250 mm3)0.5 pmol/g mouse (i.v.)[38]
MDA-MB-231Anti-EGFR-gold nanorod conjugatesBALB/c nude mice (xenograft)/Histological analysisBeclin-1
LC3
Enhanced beclin-1 and LC3 expression0.5 pmol/g mouse (i.v.)[38]
Abbreviations: ---, Not analyzed; Akt, Protein Kinase B; Atg5, Autophagy Related 5 Protein; EGFR, Epidermal Growth Factor Receptor; i.v., intravenous injection; LC3, Microtubule-Associated Protein 1A/1B-Light Chain 3; mTOR, Mammalian Target of Rapamycin; p62, Nucleoporin p62; TEM, Transmission Electron Microscopy.
Table 5. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inhibiting migration, invasion and/or metastasis.
Table 5. Non-platinum metal based complexes (NPMBC) inhibit the in vitro and in vivo growth of triple-negative breast cancer (TNBC) cells by inhibiting migration, invasion and/or metastasis.
In Vitro
Cell LineNPMBCAssayMode of Action/Pathway InvolvedEffect ObservedDoses AssayedReference
MDA-MB-231Chitosan layered gold nanorodsTranswell assay---Inhibition of cell migration50 nM[43]
MDA-MB-231Micellar gold nanoparticles conjugated with ZD6474Boyden chamber assay---~90% inhibition of cell invasion and migration1 µM[44]
MDA-MB-231Copper oxide nanowire conjugated with folic acidWound healing assay---85% inhibition of cell migration1.25 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidTranswell migration assay---80% inhibition of cell migration1.25 µg/mL[51]
MDA-MB-231Copper oxide nanowire conjugated with folic acidImmunocytochemistryFAK
PTEN
Decreased FAK expression
Up-regulated PTEN expression
1.25 µg/mL[51]
MDA-MB-231Ruthenium-based complexesTranswell assay---90.8% inhibition of cell invasion 4 µM[58]
MDA-MB-231Ruthenium-based complexesWound healing assay---79% inhibition of cell migration4 µM[58]
MDA-MB-231Ruthenium-based complexesAdhesion assayType I collagen
Fibronectin
Laminin
Vitronectin
35–70% inhibition of cancer cell adhesion to extracellular matrix proteins 8 µ[58]
MDA-MB-231Ruthenium-based complexesWound healing assay---Inhibition of cell migration 5, 10 and 20 µM[59]
MDA-MB-231Ruthenium-based complexesTranswell assay---80% inhibition of cell invasion20 µM[59]
MDA-MB-231Ruthenium-based complexesZymography assayMMP-960% of inhibition of MMP-9 expression20 µM[59]
MDA-MB-231Ruthenium-based complexesAdhesion assayType I collagen
Fibronectin
Laminin
Vitronectin
50–65% inhibition of cancer cell adhesion to type I collagen, fibronectin, laminin, vitronectin40 µM[59]
MDA-MB-231
4T1
Supermagnetic iron oxide nanoparticles conjugated with doxorubicinWound healing assay---Minimal closure of the scratch520 µM[62]
MDA-MB-231Doxorubicin-hyaluronan conjugated iron oxide nanoparticlesConfocal microscopyVimentin
e-cadherin
Decreased vimentin expression
No change in e-cadherin expression
2 µM[65]
MDA-MB-231Doxorubicin-hyaluronan conjugated iron oxide nanoparticlesELISAIL-6
IL-10
Decreased IL-6 secretion
No change in secretion of IL-10
2 µM[65]
MDA-MB-231Doxorubicin-hyaluronan conjugated iron oxide nanoparticlesSteady-Glo luciferase reagent assayNF-κBReduced NF-κB transcriptional activity (~40%)2 µM[65]
In Vivo
Cell LineNPMBCAnimal (Model)/Analysis from Animal TissueMode of Action/Pathway InvolvedEffect ObservedDoses Assayed (Route of Administration)Reference
T41Iron oxide nanoparticlesBALB/c immune-competent mice (xenograft)---Lower lung metastatic nodules than control (13 vs. 42) 20 mg iron/kg body weight (i.v.)[61]
SUM159Iron oxide nanoparticlesNOD/SCID immune-compromised mice (orthotopic)---Lower incidence of lymph node metastasis than control (25% vs. 62.5%)20 mg iron/kg body weight (i.v.)[61]
4T1Supermagnetic iron oxide nanoparticles conjugated with doxorubicinBALB/c mice (orthotopic)αvβ3Lower metastatic colonies than control (~<1 vs. ~10 at lung respectively; ~1 vs. ~3 at liver respectively) which could be related with the inhibition of αvβ30.25 mg iron/100 mm3 (i.t.)[62]
Abbreviations: ---, Not analyzed; ELISA, Enzyme-Linked ImmunoSorbent Assay; FAK, Focal Adhesion Kinase; IL, Interleukin; i.t., intratumoral injection; i.v., intravenous injection; MMP, Matrix Metalloproteinases; NF-κB, Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells; PKM, Pyruvate Kinase Isozymes; PTEN, Phosphatase and Tensin Homolog.

Share and Cite

MDPI and ACS Style

Domínguez-Martís, E.M.; Mosteiro-Miguéns, D.G.; Vigo-Gendre, L.; López-Ares, D.; Freire-Garabal, M.; Núñez-Iglesias, M.J.; Novío, S. Non-Platinum Metal Complexes as Potential Anti-Triple Negative Breast Cancer Agents. Crystals 2018, 8, 369. https://doi.org/10.3390/cryst8100369

AMA Style

Domínguez-Martís EM, Mosteiro-Miguéns DG, Vigo-Gendre L, López-Ares D, Freire-Garabal M, Núñez-Iglesias MJ, Novío S. Non-Platinum Metal Complexes as Potential Anti-Triple Negative Breast Cancer Agents. Crystals. 2018; 8(10):369. https://doi.org/10.3390/cryst8100369

Chicago/Turabian Style

Domínguez-Martís, Eva María, Diego Gabriel Mosteiro-Miguéns, Lucía Vigo-Gendre, David López-Ares, Manuel Freire-Garabal, María Jesús Núñez-Iglesias, and Silvia Novío. 2018. "Non-Platinum Metal Complexes as Potential Anti-Triple Negative Breast Cancer Agents" Crystals 8, no. 10: 369. https://doi.org/10.3390/cryst8100369

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