Skip to main content

Current shreds of evidence on the anticancer role of EGCG in triple negative breast cancer: an update of the current state of knowledge

Abstract

Triple-Negative Breast Cancer (TNBC), represents a subtype of breast cancer in which the estrogens receptor (ER) negative, the progesterone receptor (PR) negative and the human epidermal growth factor receptor 2 (HER2) negative, are not expressed. Thusly, TNBC does not respond to hormonal therapies or to those targeting the HER2 protein receptors. To overcome this flawed issue, new alternative therapies based on the use of natural substances, as the (−) - epigallocatechin 3-gallate (EGCG), has been proposed. It is largely documented that EGCG, the principal constituent of green tea, has suppressive effects on different types of cancer, including breast cancer, through the regulation of different signaling pathways. Thus, is reasonable to assume that EGCG could be viewed as a therapeutic option for the prevention and the treatment of TNBC. Here, we summarizing these promising results with the scope of turn a light on the potential roles of EGCG in the treatment of TNBC patients.

Background

Triple-negative breast cancer (TNBC), accounting for 15–20% of breast cancer with diagnosis, is an aggressive disorder with a poor prognosis frequently founded in African women with mutations in BCRA1 gene [1]. This tumor, classified as basal-like cancer on the basis of its morphology [2], does not express the estrogen receptor (ER), the progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) negative (neu) markers [3]. Thus, TNBC fails to respond to therapies targeting the HER2 protein receptors or to conventional treatments. Regarding eziopathogenesis of breast cancer, a recent study highlighted a link between the bovine leukemia virus (BLV) and breast cancer, suggesting that the DNA of BLV in mammary tissue could be considered an important marker for breast cancer [4]. Moreover, Banerjee S et al., identified the microbial signatures associated with TNBC, by using a pan-pathogen array technology in order to provide the new diagnostic potential for this type of cancer viruses [5]. Unfortunately, there are no successful therapies for TNBC, thus new elective treatments engineered on the use of natural substances, have been developed in order to change the conventional schedule of therapies. Particularly, several studies identified the polyphenols as adjuvants to chemotherapeutic drugs in TNBC cells [6, 7]. Many reports highlighted the role of EGCG against the progression of different types of cancer, including breast cancer, thought the modulation of different molecular pathways [8,9,10,11,12,13,14,15]. Specifically, it has been showed that EGCG inhibited breast cancer cell growth by enhancing the chemotherapeutic-induced cellular apoptosis [16], and by modulating different molecular signaling pathways as the nuclear factor-κB (NF- κβ), the epidermal growth factor receptor (EGFR), the mitogen-activated protein kinase (MAPK) and the phosphatidylinositol-3 (PI3) kinase [17,18,19,20]. Moreover, it has been proved that EGCG retarded the growth of MDA-MB-231 human breast cancer cells, by inactivating the β-catenin signaling pathway (Fig. 1) [21]. Overall, these findings demonstrate that EGCG could be considered as a new agent for the treatment of patients suffering from TNBC.

Fig. 1
figure 1

Molecular mechanism underlying the antitumor effect of EGCG in Triple Negative breast cancer cells. EGCG: epigallocatechin-3-gallate; PI3K: phosphatidylinositol-3 kinase; EGRF: epidermal growth factor receptor; MARK: mitogen-activated protein kinase; TNF: tumor necrosis factor; ROS: reactive oxygen species; VEGF: vascular endothelial growth factor; FAS: fas cell surface death receptor; CASP1: caspase 1; CASP3: caspase 23; CASP4: caspase 4; TP53: tumor protein p-53; TNBC: triple negative

An overview of the role of EGCG in TNBC cells growth: findings from in vitro studies

Accumulated in vitro studies highlighted the role of EGCG in the inhibition of TNBC cells growth, through the regulation of various molecular pathways (Table 1). The first study on this issue was conducted by Braicu et al. on Hs578T cells. Authors showed that the galloylated flavan-3-ols, and particularly EGCG, inhibited the cell proliferation by inducing the apoptosis, due to their ability to scavenge reactive oxygen species (ROS). Moreover, flavan-3-ols showed antioxidants or pro-oxidants properties depending on the dose and the exposure time of the cell culture [22]. Particularly, it was demonstrated that EGCG reduced or increased the concentration of reactive oxygen species (ROS) respect to controls and to others galloylated flavan-3-oils, at doses of 10 and 100 μM. In a subsequent investigation, the same authors demonstrated that EGCG (20 μM) inhibited the proliferation of Hs578T cells after 48 and 72 h of treatments, mainly by activating the apoptotic processes. By using the PCR-array technology, it has been shown that EGCG altered the expression of many genes involved in different molecular pathways (Table 1). EGCG downregulated the expression of several anti-apoptotic genes (insulin-like growth factor-1 receptor: IGF1R; induced myeloid leukemia cell differentiation protein: MCL1) while increased the expression of other genes (B-cell lymphoma 2: Bcl2; associated athanogene 3: BAG3; receptor-interacting serine/threonine-protein kinase 2: RIPK2; X-Linked Inhibitor of Apoptosis: XIAP) probably due to resistance to cancer treatment [23]. Later on, a suppressive role of EGCG on TNBC cell migration was associated with VEGF expression inhibition [24], suggesting that EGCG could be used to arrest breast tumor invasion. In a fascinating study, Braicu et al. proposed that the combination of p53 siRNA and EGCG, through the activation of apoptosis and autophagy, potentiated the antitumor effects of EGCG in Hs578T cells [25].

Table 1 In vitro findings on the role of EGCG in TNBC cells

A different molecular mechanism underlying the inhibitory role of EGCG on TNBC cell growth was reported by Hong et al. [21]. Specifically, the authors firstly evaluated a presumable association between β-catenin expression and clinical conditions of female patients with invasive ductal carcinoma, and then they examined the effect of EGCG on β- catenin expression in triple-negative breast cancer cells, MDA-MB-231. Results of western blot analysis conducted in breast cancer and normal tissue of female enrolled in this study, revealed that the expression levels of β-catenin were higher in breast cancer tissue than in normal tissue. Accordingly, EGCG decreased the cell viability of MDA-MB-231 cells in a dose-dependent manner, by downregulating the expression of β-catenin, cyclin D1, and phosphorylated Akt (p-AKT). Moreover, pre-treatment of MDA-MB-231 cells with peptidase inhibitor 3 (PI3) kinase inhibitors, (LY294002 or wortmannin) potentiated the suppressive effect of EGCG, added after 24 h, on β-catenin expression. Taken together, these data suggest that EGCG is able to inhibit the growth of MDA-MB-231 by interfering with the β-catenin signaling pathway.

Encouraging results on the effects of (−)-Epigallocatechin 3-gallate synthetic analogs on TNBC cells were depicted by Crous-Masò. In this study were generated three diesters (G28, G37, G56), two G28 derivatives and two monoesters M1 and M2. Then, these compounds were tested in vitro on MDA-MB-231 cells. Specifically were evaluated their cytotoxic effects, their inhibition of lipogenic enzyme fatty acid synthase (FANS) and their apoptotic activities. Data emerged from these experiments, showed that all compounds blocked the activity of FASN. Particularly, the monoesters inhibited the growth of MDA-MB-231 cells more than the diesters, and only the monoesters induced the apoptosis detected by poly (ADP-ribose) polymerase (PARP) cleavage. Altogether, these results suggest that these polyphenolic compounds, particularly the monoesters, due to their inhibitory role on FASN, could represent a new strategy for TNBC treatment [26].

A recent study evaluated the effects of Suberoylanilide hydroxamic acid (SAHA) histone deacetylase (HDAC) inhibitor and EGCG on the growth and the proliferation of TNBC cells [27]. Results showed that these substances reduced the expression of miR-221/222 and N-cadherin and increased the expression of p27, phosphatase and tensin homolog (PTEN) estrogen receptor alpha (ERα) and E-cadherin. These findings, likewise associated with reduced activity of DNA methyltransferase (DNMTs), indicate that SAHA and EGCG reduced the growth and the proliferation of TNBC cancer cells, probably by acting on epigenetic mechanisms.

The antitumor effects of EGCG in TNBC mouse models: a current state of knowledge

The antitumor effects of EGCG on TNBC have been also confirmed by in vivo experiments, as summarized in Table 2. A first study was conducted by Thangapazham al [20]. Authors showed that EGCG (administered at the dose of 1 mg/animal in 100 μl of distilled water for 10 weeks), and GTP (green tea polyphenols, 1% for 10 weeks) decreased the proliferation and increased the apoptosis of tumors of TNBC mouse model. Importantly, these results were firstly proved in vitro. Specifically, GTP (10–150 μg/ml) and EGCG (1–200 μg) inhibited cell growth in a dose-dependent manner. Moreover, an arrest of the cell cycle at G1 phase, confirmed by a downregulated expression of Cyclin D, Cyclin E, Cyclin-dependent kinase 4 (CDK 4), Cyclin-dependent kinase 1 (CDK 1) and proliferating cell nuclear antigen (PCNA), was observed in cells treated with GTP and EGCG. An interesting study conducted on the same TNBC mouse model tested the effect of a novel EGCG prodrug on tumor growth [28]. Specifically, this compound, named Pro-EGCG (1), was engineered to enhance the stability of EGCG by introducing the peracetate-protecting groups to the reactive hydroxyls of EGCG. By using this approach, the authors showed that in MDA-MB-231 cells, Pro-EGCG (1) was converted to its parent compound EGCG that was then accumulated. Thus, the cells treated with Pro-EGCG (1) (50 μmol/L), showed increased levels of proteasome inhibition, growth suppression and apoptosis compared to cells treated with EGCG. These data were also confirmed in vivo by injecting the MDA-MB-231 cells in nude mice, followed by treatment with Pro-EGCG (1) (50 mg/kg) or EGCG (50 mg/kg) for 31 days. Data emerged from this experiment showed that Pro-EGCG (1) inhibited breast tumor growth and proteasome, while induced the apoptosis compared to EGCG, suggesting its use for TNBC prevention and treatment. Similar reports were showed by Yang et al. [29] with the utilization of the prodrugs of EGCG’s flour-substituted analogs: Pro-F-EGCG2 (Pro-F2) or Pro-F-EGCG4 (Pro-F4) and Pro-EGCG 1. These compounds (50 mg/kg administered by s.c. injection daily for 31 days), equally, inhibited the growth and the proteasome and enhanced the apoptosis in breast tumors of mice.

Table 2 In vivo experiments on the antitumor effects of EGCG on TNBC: an update

Limitations of the use of EGCG into clinical practice and future perspectives

The aforementioned results strongly indicate that EGCG can restrain the growth and the proliferation of TNBC by acting on different molecular pathways. Thus, EGCG might be viewed as an alternative treatment for TNBC patients. Notwithstanding these encouraging pre-clinical outcomes, no clinical trials have been conducted, until now, on TNBC patients. Lamentably, the data obtained from in vivo and human epidemiological studies on other types of breast cancer, are inconsistent and in some cases contradictory [30]. Moreover, EGCG possesses poor bioavailability and poor stability and these highlights change between in vitro and in vivo conditions [31, 32], thus representing a limitation for EGCG’s use in human populations. To conquer this issue, new strategies based on the use of nanoparticles (NPs) or micro-particles in which EGCG is encapsulated, have been effectively built to improve EGCG’s stability and bioavailability [33,34,35,36,37,38,39].

Further investigations will be important not only to improve EGCG oral bioavailability, yet additionally to stabilize this compound in the stomach related tract. Overall, EGCG could be firmly viewed as a powerful inhibitor of TNBC progression.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

BAD:

BCL2 associated agonist of cell death

BAG3:

bcl2 associated athanogene 3

BAK1:

BCL2 antagonist/killer 1

BRAF:

b-raf serine/threonine-protein

CARD6:

Caspase recruitment domain family member 6

CASP1:

Caspase 1

CASP3:

Caspase 3

CASP4:

Caspase 4

EGCG:

Epigallocatechin-3-gallate

ERα:

Estrogen receptor alpha

FAS:

Fas cell surface death receptor

GTP:

Green tea polyphenols

IGF1R:

Insulin-like growth factor-1 receptor

LTBR:

Lymphotoxin beta receptor

MCL1:

Induced myeloid leukemia cell differentiation protein

miR-221/222:

MicroRNA 221/222

p-AKT:

Phospho-AKT

PARP:

Poly(ADP- ribose) polymerase

Pro-EGCG(1):

Peracetate-protecting groups to the reactive hydroxyls of (−)-EGCG

Pro-F2:

Prodrugs of fluoro-substituted EGCG analog-42

Pro-F4:

Prodrugs of fluoro- substituted EGCG analog-4

PTEN:

Phosphatase and tensin homolog

PYCARD:

Apoptosis-associated speck-like protein containing a card

RIPK2:

Receptor-interacting serine/threonine-protein kinase 2

ROS:

Reactive oxygen species

SAHA:

Suberoylanilide hydroxamic acid

TNBC:

Triple-negative breast cancer

TNF:

Tumor necrosis factor

TNFRSF1A:

Tumor necrosis factor receptor superfamily member 1A

TNFRSF25:

Tumor necrosis factor receptor superfamily member 25

TNFSF8:

Tumor necrosis factor ligand superfamily, member 8

TP53:

Tumor protein p-53

VEGF:

Vascular endothelial growth factor

XIAP:

X-linked inhibitor of apoptosis protein

References

  1. Alluri P, Newman L. Basal-like and triple negative breast cancers: searching for positives among many negatives. Surg Oncol Clin N Am. 2014;23:567–77. https://0-doi-org.brum.beds.ac.uk/10.1016/j.soc.2014.03.003.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P, Narod SA. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007;13:4429–34.

    Article  Google Scholar 

  3. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13:674–90.

    Article  CAS  Google Scholar 

  4. Buehring GC, et al. Bovine leukemia virus DNA in human breast tissue. Emerg Infect Dis. 2014;20:772–82. https://0-doi-org.brum.beds.ac.uk/10.3201/eid2005.131298.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Banerjee S, Wei Z, Tan F, Peck KN, Shih N, Feldman M, Rebbeck TR, Alwine JC, Robertson ES. Distinct microbiological signatures associated with triple negative breast cancer. Sci Rep. 2015;15:15162. https://0-doi-org.brum.beds.ac.uk/10.1038/srep15162.

    Article  CAS  Google Scholar 

  6. Chisholm K, Bray BJ, Rosengren RJ. Tamoxifen and epigallocatechin gallate are synergistically cytotoxic to MDA-MB-231 human breast cancer cells. Anti-Cancer Drugs. 2004;15:889–97.

    Article  CAS  Google Scholar 

  7. Sak K. Chemotherapy and dietary phytochemical agents. Chemother Res Pract. 2012;2012:282570.

    PubMed  PubMed Central  Google Scholar 

  8. Bimonte S, Albino V, Piccirillo M, et al. Epigallocatechin-3-gallate in the prevention and treatment of hepatocellular carcinoma: experimental findings and translational perspectives. Drug Des Devel Ther. 2019;13:611–21. https://0-doi-org.brum.beds.ac.uk/10.2147/DDDT.S180079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bimonte S, Leongito M, Barbieri A, et al. Inhibitory effect of (−)-epigallocatechin-3-gallate and bleomycin on human pancreatic cancer MiaPaca-2 cell growth. Infect Agent Cancer. 2015;10:22. https://0-doi-org.brum.beds.ac.uk/10.1186/s13027-015-0016-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Modernelli A, Naponelli V, Giovanna Troglio M, et al. EGCG antagonizes Bortezomib cytotoxicity in prostate cancer cells by an autophagic mechanism. Sci Rep. 2015;5:15270.

    Article  CAS  Google Scholar 

  11. Avtanski D, Poretsky L. Phyto-polyphenols as potential inhibitors of breast cancer metastasis. Mol Med. 2018;24(1):29. https://0-doi-org.brum.beds.ac.uk/10.1186/s10020-018-0032-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yamada S, Tsukamoto S, Huang Y, Makio A, Kumazoe M, Yamashita S, Tachibana H. Epigallocatechin-3-O-gallate up-regulates microrna-let-7b expression by activating 67-kda laminin receptor signaling in melanoma cells. Sci Rep. 2016;6:19225. https://0-doi-org.brum.beds.ac.uk/10.1038/srep19225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. He L, Zhang E, Shi J, Li X, Zhou K, Zhang, Le AD, Tang X. (−)-epigallocatechin-3-gallate inhibits human papillomavirus (hpv)-16 oncoprotein-induced angiogenesis in non-small cell lung cancer cells by targeting hif-1alpha. Cancer Chemother Pharmacol. 2013;71:713–25. https://0-doi-org.brum.beds.ac.uk/10.1007/s00280-012-2063-z.

    Article  CAS  PubMed  Google Scholar 

  14. Lee SH, Nam HJ, Kang HJ, Kwon HW, Lim YC. Epigallocatechin-3-gallate attenuates head and neck cancer stem cell traits through suppression of notch pathway. Eur J Cancer. 2013;49:3210–8. https://0-doi-org.brum.beds.ac.uk/10.1016/j.ejca.2013.06.025.

    Article  CAS  PubMed  Google Scholar 

  15. Adachi S, Nagao T, Ingolfsson HI, Maxfield FR, Andersen OS, Kopelovich L, Weinstein IB. The inhibitory effect of (−)-epigallocatechin gallate on activation of the epidermal growth factor receptor is associated with altered lipid order in ht29 colon cancer cells. Cancer Res. 2007;67:6493–501. https://0-doi-org.brum.beds.ac.uk/10.1158/0008-5472.

    Article  PubMed  Google Scholar 

  16. Roy AM, Baliga MS, Katiyar SK. Epigallocatechin-3-gallate induces apoptosis in estrogen receptor-negative human breast carcinoma cells via modulation in protein expression of p53 and Bax and caspase-3 activation. Mol Cancer Ther. 2005;4:81–90.

    CAS  PubMed  Google Scholar 

  17. Hsu YC, Liou YM. The anticancer effects of (−)-Epigalocathine-3-gallate on the signaling pathways associated with membrane receptors in MCF-7 cells. J Cell Physiol. 2011;226:2721–30.

    Article  CAS  Google Scholar 

  18. Khan N, Afaq F, Saleem M, Ahmad N, Mukhtar H. Targeting multiple signaling pathways by green tea polyphenol (−)-epigallocatechin-3-gallate. Cancer Res. 2006;66:2500–5.

    Article  CAS  Google Scholar 

  19. Stuart EC, Scandlyn MJ, Rosengren RJ. Role of epigallocatechin gallate (EGCG) in the treatment of breast and prostate cancer. Life Sci. 2006;79:2329–36.

    Article  CAS  Google Scholar 

  20. Thangapazham RL, Singh AK, Sharma A, Warren J, Gaddipati JP, Maheshwari RK. Green tea polyphenols and its constituent epigallocatechin gallate inhibits proliferation of human breast cancer cells in vitro and in vivo. Cancer Lett. 2007;245:232–41.

    Article  CAS  Google Scholar 

  21. Hong OY, Noh EM, Jang HY, et al. Epigallocatechin gallate inhibits the growth of MDA- MB-231 breast cancer cells via inactivation of the β-catenin signaling pathway. Oncol Lett. 2017;14(1):441–6.

    Article  CAS  Google Scholar 

  22. Braicu C, Pilecki V, Balacescu O, Irimie A, Neagoe IB. The relationships between biological activities and structure of flavan-3-ols. Int J Mol Sci. 2011;12(12):9342–53.

    Article  CAS  Google Scholar 

  23. Braicu C, Gherman C. Epigallocatechin gallate induce cell death and apoptosis in triple negative breast cancer cells Hs578T. J Drug Target. 2012;21(3):250-256. https://0-doi-org.brum.beds.ac.uk/10.3109/1061186X.2012.740673. Epub 2012 Nov 19.

    Article  Google Scholar 

  24. Braicu C, Gherman CD, Irimie A, Berindan-Neagoe I. Epigallocatechin-3-Gallate (EGCG) inhibits cell proliferation and migratory behaviour of triple negative breast cancer cells. J Nanosci Nanotechnol. 2013;13(1):632–7.

    Article  CAS  Google Scholar 

  25. Braicu C, Pileczki V, Pop L, et al. Dual targeted therapy with p53 siRNA and Epigallocatechingallate in a triple negative breast cancer cell model. PLoS One. 2015;10(4):e0120936.

    Article  Google Scholar 

  26. Crous-Masó J, Palomeras S, Relat J, et al. (−)-epigallocatechin 3-gallate synthetic analogues inhibit fatty acid synthase and show anticancer activity in triple negative breast cancer. Molecules. 2018;23(5):1160.

    Article  Google Scholar 

  27. Lewis KA, Jordan HR, Tollefsbol TO. Effects of SAHA and EGCG on growth potentiation of triple-negative breast cancer cells. Cancers (Basel). 2018;11(1):23.

    Article  Google Scholar 

  28. Landis-Piwowar KR, Huo C, Chen D, Milacic V, Shi G, Chan TH, Dou QP. A novel prodrug of the green tea polyphenol (−)-epigallocatechin-3-gallate as a potential anticancer agent. Cancer Res. 2007;67(9):4303–10.

    Article  CAS  Google Scholar 

  29. Yang H, Sun DK, Chen D, et al. Antitumor activity of novel fluoro-substituted (−)- epigallocatechin-3-gallate analogs. Cancer Lett. 2009;292(1):48–53.

    Article  Google Scholar 

  30. Xiang LP, Wang A, Ye JH, et al. Suppressive effects of tea catechins on breast cancer. Nutrients. 2016;8(8):458 Published 2016 Jul 28.

    Article  Google Scholar 

  31. Weng Z, Greenhaw J, Salminen WF, Shi Q. Mechanisms for epigallocatechin gallate induced inhibition of drug metabolizing enzymes in rat liver microsomes. Toxicol Lett. 2012;214:328–38.

    Article  CAS  Google Scholar 

  32. Lorenz M, Paul F, Moobed M, Baumann G, Zimmermann BF, Stangl K, Stangl V. The activity of catechol-O-methyltransferase (COMT) is not impaired by high doses of epigallocatechin-3-gallate (EGCG) in vivo. Eur J Pharmacol. 2014;740:645–51.

    Article  CAS  Google Scholar 

  33. Suganuma M, Okabe S, Sueoka N, Sueoka E, Matsuyama S, Imai K, Nakachi K, Fujiki H. Green tea and cancer chemoprevention. Mutat Res. 1999;428:339–44.

    Article  CAS  Google Scholar 

  34. Forester SC, Lambert JD. The catechol-O-methyltransferase inhibitor, tolcapone, increases the bioavailability of unmethylated (−)-epigallocatechin-3-gallate in mice. J Funct Foods. 2015;17:183–8.

    Article  CAS  Google Scholar 

  35. Krupkova O, Ferguson SJ, Wuertz-Kozak K. Stability of (−)-epigallocatechin gallate and its activity in liquid formulations and delivery systems. J Nutr Biochem. 2016;37:1–12.

    Article  CAS  Google Scholar 

  36. Dube A, Nicolazzo JA, Larson I. Chitosan nanoparticles enhance the intestinal absorption of the green tea catechins (+)-catechin and (−)-epigallocatechingallate. Eur J Pharm Sci. 2014;41:219–25.

    Article  Google Scholar 

  37. Yadav R, Kumar D, Kumari A, Yadav SK. Encapsulation of catechin and epicatechin on BSA NPs improved their stability and antioxidant potential. EXCLI J. 2014;13:331–46.

    PubMed  PubMed Central  Google Scholar 

  38. Liang J, Cao L, Zhang L, Wan X. Preparation, characterization, and in vitro antitumor activity of folate conjugated chitosan coated EGCG nanoparticles. Food Sci Biotechnol. 2014;23:569–75.

    Article  CAS  Google Scholar 

  39. Fangueiro JF, Calpena AC, Clares B, Andreani T, Egea MA, Veiga FJ, Garcia ML, Silva AM, Souto EB. Biopharmaceutical evaluation of epigallocatechin gallate-loaded cationic lipid nanoparticles (EGCG-LNs): in vivo, in vitro and ex vivo studies. Int J Pharm. 2016;502:161–9.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

The authors are grateful to Alessandra Trocino and Mrs. Maria Cristina Romano from the National Cancer Institute of Naples for providing excellent bibliographic service and assistance.

Author information

Authors and Affiliations

Authors

Contributions

The present review was mainly written by SB and MC. All authors contributed toward data analysis, drafting and critically revising the paper, gave final approval of the version to be published, and agree to be accountable for all aspects of the work.

Corresponding author

Correspondence to Sabrina Bimonte.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bimonte, S., Cascella, M., Barbieri, A. et al. Current shreds of evidence on the anticancer role of EGCG in triple negative breast cancer: an update of the current state of knowledge. Infect Agents Cancer 15, 2 (2020). https://0-doi-org.brum.beds.ac.uk/10.1186/s13027-020-0270-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13027-020-0270-5

Keywords