Muhammad Younasa,
Christophe Hanob,
Nathalie Giglioli-Guivarc'hc and
Bilal Haider Abbasi*abc
aDepartment of Biotechnology, Quaid-i-Azam University, Islamabad-45320, Pakistan. E-mail: bhabbasi@qau.edu.pk; Fax: +92-51-90644121; Tel: +92-51-90644121 Tel: +33-767-97-0619
bLaboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans, F 28000 Chartres, France
cEA2106 Biomolecules et Biotechnologies Vegetales, Universite Francois-Rabelais de Tours, Tours, France
First published on 22nd August 2018
Breast cancer is one of the most commonly diagnosed cancers around the globe and accounts for a large proportion of fatalities in women. Despite the advancement in therapeutic and diagnostic procedures, breast cancer still represents a major challenge. Current anti-breast cancer approaches include surgical removal, radiotherapy, hormonal therapy and the use of various chemotherapeutic drugs. However, drug resistance, associated serious adverse effects, metastasis and recurrence complications still need to be resolved which demand safe and alternative strategies. In this scenario, phytochemicals have recently gained huge attention due to their safety profile and cost-effectiveness. These phytochemicals modulate various genes, gene products and signalling pathways, thereby inhibiting breast cancer cell proliferation, invasion, angiogenesis and metastasis and inducing apoptosis. Moreover, they also target breast cancer stem cells and overcome drug resistance problems in breast carcinomas. Phytochemicals as adjuvants with chemotherapeutic drugs have greatly enhanced their therapeutic efficacy. This review focuses on the recently recognized molecular mechanisms underlying breast cancer chemoprevention with the use of phytochemicals such as curcumin, resveratrol, silibinin, genistein, epigallocatechin gallate, secoisolariciresinol, thymoquinone, kaempferol, quercetin, parthenolide, sulforaphane, ginsenosides, naringenin, isoliquiritigenin, luteolin, benzyl isothiocyanate, α-mangostin, 3,3′-diindolylmethane, pterostilbene, vinca alkaloids and apigenin.
Human beings have always been suffered from infections by fungi, parasites, bacteria, viruses and many health disorders such as pain, inflammation, digestive complications and colds etc.12 Recent medicines, based on antibiotics and synthetic drugs, have come into practice during the previous 150 years. Before that, humans had to depend on drugs derived from plants, animals and fungi. The curing of health disorders and infections with herbal medicines includes active natural products, known as secondary metabolites or phytochemicals, which are found in almost all plant species.12 Secondary metabolites encompass a diverse group of organic compounds which have a vital role in plant defence systems and assist the interaction with the biotic environment. Many secondary metabolites like phenolics, alkaloids and terpenes are classified on the basis of their biosynthetic origin which possess different biological properties and are employed as pharmaceuticals, flavors, colors, agrochemicals, fragrances, food additives and biopesticides.13–15 Secondary metabolites specifically modulate a molecular target in humans or animals. Such targets often include neuroreceptors, ion pumps, ion channels and elements of the cytoskeleton or enzymes degrading neurotransmitters.12,16–20 An estimated more than 5000 distinct phytochemicals have been recognized in vegetables, fruits and grains, however, a large proportion is yet to be identified and understood before we can completely comprehend their health benefits in whole foods.21 Phytochemicals can be categorized into different classes such as phenolics, terpenes, organosulfur compounds and alkaloids (Fig. 1).393,394 The molecular structures of various phytochemicals involved in breast cancer chemoprevention with their major plant sources are given in Table 1.
Phytochemicals display diverse range of disease-preventing or protective effects. They have been used since ancient times to cope with various ailments including cancer, diabetes, cardiovascular diseases, inflammation, neurological disorders and skin diseases etc.22–28 Numerous epidemiological studies have predicted reduced cancer incidence with the use of phytochemicals.29–31 Phytochemicals such as curcumin,32 resveratrol,33,34 epigallocatechin gallate (EGCG),35,36 silibinin,37 benzyl isothiocyanate,38,39 genistein,40 kaempferol,41,42 thymoquinone,43 quercetin,44 parthenolide,45 sulforaphane,46 naringenin,47,327,330 isoliquiritigenin48,49 and ginsenosides50–52 have been shown to suppress breast carcinoma via modulation of various signalling transduction pathways, genes and gene products. These phytochemicals exert anti-breast cancer effects by inducing cellular apoptosis and reducing cell proliferation through modulation of various targets (Fig. 2 & 4). Additionally, phytochemicals inhibit the angiogenesis, metastasis and migratory behaviours in breast cancer cells (Fig. 3 & 4). Moreover, these compounds greatly enhanced the therapeutic efficacy of different anti-cancer drugs, overcame drugs resistance in breast cancer cells and also achieved sensitization to radiations.53–58 The compounds targeted breast cancer stem cells (bCSCs)/progenitor cells.59,60 CSCs are mainly involved in promotion of invasion, metastasis, abnormal proliferation, recurrence and drug resistance.61 The self-renewal capability of CSCs is related to the regulatory pathways of Notch, Wnt/β-catenin, hedgehog and P13K/Akt. These pathways might also contribute in the maintenance of CD44+/CD24−/low bCSCs stemness.59,62 Dandawate et al. (2016) reviewed the role of phytochemicals in targeting bCSCs.60 Abdal Dayem et al. (2016) reviewed the effect of polyphenols against breast cancer and CSCs (Fig. 5).395 Petric et al. (2015) reviewed some phytochemicals modulating signalling pathways in breast and hormone related cancers.63 Moreover, Siddiqui et al. (2015) have also put some light on therapeutic effect of phytochemicals in breast cancer.64 In this review article, we summarize various phytochemicals that are involved in breast cancer prevention, by putting in view the recently recognized molecular mechanisms, which may serve useful in future drugs development.
Fig. 4 Possible number of key targets involved in breast cancer cells proliferation, apoptosis, angiogenesis, invasion and metastasis and their regulation by phytochemicals. |
Fig. 5 Polyphenols and their modes of action against breast cancer. Blue arrows indicate activation while red arrows indicate inhibition. CUR = curcumin, RSV = resveratrol, GEN = genistein, APG = apigenin, QUR = quercetin. Idea adopted from Abdal Dayem et al. (2016).395 |
Phytochemical | Cell line/model | Proposed mechanism | Effect | Ref. |
---|---|---|---|---|
Curcumin | MCF-7 | ↓GSTP1 methylation | ↑Glutathione S-transferase Pi 1 | 72 |
MDA-MB-361 | ↓Sp1 expression, ↓DLC1 methylation | ↓Growth, ↑re-expression of the tumor suppressor: deleted in liver cancer 1 | 73 | |
MCF-7 | ↓miR-21, ↑PTEN, ↑Akt, ↑caspase-3, ↑caspase-9 | ↑Apoptosis | 66 | |
MCF-7 & MCF-7/DPP | ↓CCAT1 expression, ↓PI3K, ↓p-Akt, ↓p-mTOR | ↑Autophagy, ↑sensitization to cisplatin | 80 | |
MCF-10F, MDA-MB-231 & Tumor 2 | ↓β-Catenin, ↓N-cadherin, ↓E-cadherin, ↓slug, ↓Twist1, ↓AXL, ↓fibronectin, ↓vimentin | ↓Migration, ↓invasion | 88 | |
MDA-MB-231 | ↑AMPK, ↓Akt | ↑Autophagy, ↓proliferation, ↓migration | 280 | |
MDA-MB-231 | ↓Intracellular FAS | ↑Apoptosis | 69 | |
MDA-MB-231 | ↓Slug/HK2 axis | ↑Apoptosis, ↓4-OHT resistance | 281 | |
MCF-7, MCF10A, SUM149 | ↓SCD, ↓CD49f, ↓LDH1A3, ↓TP63, ↓PROM1 | ↓bCSCs self-renewal | 282 | |
MCF-7 | ↓Cyclin B1, ↓Cdc2 | ↓Colonization | 283 | |
MCF-7 | ↓RhoA, ↓ROCK1, ↓ROCK2, ↓MMP-2, ↓MMP-9 | ↓Invasion | 83 | |
MDA-MB-231 | ↓Cyclin D1, ↓p65, ↓PECAM-1 | ↓Angiogenesis, ↓proliferation, ↑apoptosis | 84 | |
MDA-MB-231 | ↑miR181b, ↓CXCL1, ↓CXCL2 | ↓Metastasis, ↓proliferation, ↓invasion, ↑apoptosis | 284 | |
MDA-MB-231 & MCF-7 | ↓Bcl-2, ↑Bax | ↑Apoptosis | 68 | |
MCF-7 & T47D | ↑E-Cadherin/β-catenin negative feedback loop | ↓Migration of bCSCs | 71 | |
MCF-7 | ↑Nrf2 expression, ↓Fen1 expression | ↓Proliferation | 70 | |
MCF-7 & MDA-MB-231 | ↓NF-κB-Snail signalling pathway, ↑E-cadherin, ↓vimentin | ↓Invasion | 285 | |
MCF-7/LCC2 & MCF-7/LCC9 | ↓NF-κB, ↓Src, ↓FAK, ↓Akt, ↓mTOR, ↓EZH2, ↓Bcl-2, ↓Bcl-xL, ↓cyclin D1, ↓c-Myc, ↑ERK1/2 | ↓Proliferation, ↑apoptosis, ↓endocrine resistance, ↑sensitization to tamoxifen | 286 | |
MCF-7 | ↓uPA expression, ↓NF-κB | ↓Adhesion, ↓invasion | 82 | |
MCF-7, MCF-10A, MDA-MB-231, SK-BR-3h | ↓HER-2 oncoprotein, ↓MAPK, ↓p-Akt, ↓NF-κB | ↓Proliferation, ↓migration | 287 | |
MCF-7 & MDA-MB-231 | ↓DNMT1, ↓RASSF1A methylation | ↑ RASSF1A | 74 | |
MDA-MB-231 | ↓pERK1/2, ↓pEGFR | ↑Apoptosis, ↓proliferation | 288 | |
MCF-7 | ↓MAPK, ↓PKC, ↓NF-κB | ↓Invasion | 289 | |
EGCG | In vivo | ↓Ki-67 | ↓Proliferation | 290 |
T-47D | ↓Telomerase, ↓PI3K/AKT | ↑Apoptosis | 92 | |
MCF-7 | ↓Bcl-2, ↑p53 | ↑Apoptosis, ↓proliferation | 93 | |
MDA-MB-231 | ↓β-Catenin signalling pathway | ↓Proliferation | 94 | |
T-47D | ↓ERα protein levels | ↓Proliferation | 291 | |
MCF-7/TAM | ↓Nrf2 expression | ↓Tamoxifen resistance | 100 | |
MCF-7/DOX | ↓MMP-2, ↓MMP-9 activity | ↓Doxorubicin resistance | 101 | |
MDA-MB-231 & MDA-MB-436 | ↓ER-α36, ↓MAPK/ERK, ↓EGFR, ↓PI3K/AKT | ↓Proliferation, ↓bCSCs growth | 98 | |
MCF-7, T-47D & SK-BR-3 | ↓Hsp90/PR-B/HDAC interactions, ↑p38/CK2, ↓ERα | ↓Proliferation, ↓colonization | 96 | |
MCF-7 & MDA-MB-231 | ↓H3K9/18 acetylation, ↓EZH2, ↓class I HDAC, ↓TIMP-3, ↓H3K2 trimethylation | ↓Progression & invasion | 292 | |
SUM-149 & SUM-190 | ↓ALDH+ cells, ↓VEGF-D, ↓cyclin D1, ↓RhoC, ↓FN1, ↓E-cadherin, ↓Bcl-XL, ↓VIM, ↑c-PARP, ↑cleaved caspase-3 | ↓Proliferation, ↓migration, ↓invasion, ↑apoptosis, ↓lymphangiogenesis, ↓tumorsphere formation | 293 | |
Hs578T | ↓VEGF | ↓Proliferation, ↓migration & invasion | 294 | |
MCF-7 | ↓VEGF, ↓HIF-1α | ↓Proliferation | 295 | |
4T1 & RAW264.7 | ↑miR-16, ↓IKKα, ↓CSF-1, ↑IL-6, ↓CCL-2, ↑TGF-β, ↑TNF-α | ↓Tumor growth, ↓TAM infiltration, ↓M2 polarization | 296 | |
MCF-7 & MDA-MB-231 | ↑p21WAF1, ↓DNMT1, ↓HDAC1, ↓MeCP2, ↓RARβ2, ↓cyclin D2 methylation, ↓TMS1 methylation, ↓MGMT methylation | ↓Cell viability | 297 | |
Genistein | Sprague-Dawley rats | ↓UPR, ↓GRP78, ↓IRE1α, ↓Beclin-1, ↓ATF4, ↓TGFβ, ↓Foxρ3, ↑CD8a | ↓Tamoxifen resistance, ↓recurrence | 110 |
MCF-7-C3 & T-47D | ↓CIP2A, ↓E2F1 | ↑Apoptosis, ↓proliferation | 106 | |
MCF-7 | ↑miR-23b | ↑Cytotoxicity | 111 | |
MDA-MB-231 & MCF-7 | ↓DNMTI, ↑ATM, ↑PTEN, ↑APC, ↑SERPINB5 | ↓Cell viability | 108 | |
MCF-7/Adr | ↓ HER2/neu | ↑Apoptosis, ↓doxorubicin resistance | 298 | |
MCF-7 & 3T3-L1 | ↓ERα, ↓cyclin D1, ↓Bcl-2, ↑Bax | ↓Proliferation, ↑apoptosis | 103 | |
MCF-7 | ↓Hedgehog, ↓Gli1 | ↓bCSCs | 113 | |
MDA-MB-231 & MCF-7 | ↑ATM, ↑Chk2, ↑Cdc25C, ↑Cdc2, ↑Bax, ↑p53, ↓Bcl-2, ↓Rad51 | ↑Radiosensitivity, ↑apoptosis | 299 | |
MCF-7, SK-BR-3 & ZR-75-1 | ↓ERα, ↓c-erbB-2 expression | ↓Proliferation | 105 | |
MDA-MB-231 | ↓pERK1/2, ↑Bax/Bcl-2 ratio | ↑Apoptosis, ↓proliferation | 300 | |
MDA-MB-231 | ↓NF-κB, ↓cyclin B1, ↓Bcl-2, ↓Bcl-xL | ↓Proliferation, ↑apoptosis | 301 | |
MCF-7 & UACC-3199 | ↓DNMT-1, ↓cyclin D1, ↑p53, ↑BRCA-1, ↓CpG methylation, ↑CYP1A1 | ↓Proliferation | 40 | |
Resveratrol | MDA-MB-231 & MCF-7 | ↑ATP2A3, ↓Bcl-2, ↓Ki67, ↑Bcl-2L11 (BIM) | ↑Apoptosis, ↓proliferation | 114 |
T47-D | ↓p53, ↓ERα | ↓Proliferation | 302 | |
MDA-MB-231 | ↓AURKA, ↓PLK1, ↓cyclin D1, ↓cyclin B1 | ↓Cell cycle progression, ↑apoptosis, ↓viability | 117 | |
MDA-MB-231 & MDA-MB-468 | ↓YAP, ↓RhoA | ↓Invasion | 116 | |
MDA-MB-231 & MCF-7 | ↓ XIAP, ↓Bcl-2, ↑CASP-8, ↑CASP-9, modulation of miR-125b-5p, miR-409-3p, miR-200c-3, miR-542-3p & miR-122-5p | ↑Apoptosis | 128 | |
MCF-7/DOX | ↓MDR-1, ↓P-glycoprotein | ↓Doxorubicin resistance, ↓proliferation | 124 | |
MCF-7, SUM159 | ↓ Wnt, ↓β-catenin | ↓bCSCs proliferation, ↑autophagy, ↓mammospheres | 303 | |
SKBR-3 | ↓FASN, ↓HER2, ↑PEA3, ↓p-Akt, ↑PTEN, ↓PI3K, ↓Akt, ↓mTOR | ↓Proliferation, ↑apoptosis | 115 | |
MCF-7 | ↓PFK activity, ↓glucose consumption, ↓ATP content | ↓Cell viability | 304 | |
MCF-7/TR | ↓TGF-β, ↓Smad cascade, ↓EMT | ↓Tamoxifen resistance, ↑apoptosis | 126 | |
MCF-7 | ↓HSP27 | ↑Sensitization to doxorubicin | 125 | |
MCF-7 & MDA-MB-231 | ↑p53, ↓procaspase 8, ↑CASP-7, ↑CASP-9, ↑p-Chk2, ↓cyclin A, ↓Thr160-phosphorylated CDK2, ↓CDK7, ↑cell cycle arrest | ↑Sensitization to melphalan | 127 | |
Quercetin | MDA-MB-231 & MDA-MB-157 | ↓FASN, ↓β-catenin, ↓Bcl-2, ↑caspase-3 | ↑Apoptosis, ↓proliferation | 44 |
MCF-7 | ↓Bcl-2, ↑Bax | ↑Apoptosis, ↑necroptosis | 130 | |
MCF-7 | ↓VEGF, ↓VEGFR2, ↓NFATc3, ↓calcineurin pathway | ↓Angiogenesis | 137 | |
MCF-7Ca/TAM-R | ↓Her-2, ↑ERα | ↓Proliferation, ↑apoptosis, ↓tamoxifen resistance | 134 | |
MDA-MB-231 & MDA-MB-468 | ↑E-Cadherin, ↓vimentin, ↓c-Myc, ↓cyclin D1 | ↓Metastasis, ↓proliferation | 136 | |
MCF-7 | ↓Twist, ↓p38MAPK, ↓cyclin D1, ↓p21 | ↑Apoptosis | 131 | |
MCF-7 | ↓Proteasome, ↑CASP-3, ↑CASP-7 | ↓Proliferation, ↑apoptosis | 129 | |
MCF-7 & MDA-MB-231 | ↑miR-146a, ↓EGFR, ↑Bax, ↑CASP-3 | ↓Proliferation, ↓invasion, ↑apoptosis | 138 | |
MCF-7 | ↓Survivin | ↓Proliferation | 132 | |
MCF-7/TR | ↓Cyclin E2 | ↑Sensitization to tamoxifen | 305 | |
MCF-7 | ↓Bcl-2, ↑Bax | ↓Proliferation, ↑apoptosis | 306 | |
Kaempferol | MCF-7 | ↓pIRS-1, ↓pAkt, ↓pMEK1/2, pERK1/2, ↓cyclin D1, ↓cyclin E, ↑p21, ↓cathepsin D | ↓Proliferation, ↓metastasis, ↑apoptosis | 243 |
MCF-7 | ↓Bcl-2, ↑Bax, ↑PARP cleavage | ↑Apoptosis | 242 | |
MCF-7 | ↓Cathepsin B, ↓cathepsin D, ↓N-cadherin, ↓snail, ↓slug, ↑E-cadherin | ↓Proliferation, ↓migration, ↓invasion, ↓metastasis | 307 | |
MDA-MB-231 & MDA-MB-453 | ↓RhoA, ↓Rac1 | ↓Migration, ↓invasion | 42 | |
MCF-7 | ↑E-cadherin, ↓MMP-9, ↓MMP-2, ↓cathepsin B, ↓cathepsin D, ↓N-cadherin, ↓snail, ↓slug | ↓Metastasis | 244 | |
MCF-7 | ↓GLUT1, ↓MCT1 | ↓Proliferation, ↑cytotoxicity | 241 | |
Apigenin | MDA-MB-231 | ↓Cyclin A, ↓cyclin B, ↓CDK1, ↑p21WAF1/CIP1 | ↓Proliferation | 176 |
MCF-7/ADR | ↓MDR1, ↓P-gp, ↓p-STAT3, ↓VEGF, ↓MMP-9 | ↑Apoptosis, ↓colonization, ↓adriamycin resistance | 173 | |
MDA-MB-231 | ↓TNFα, ↓CCL2, ↓GMCSF, ↓IL-1α, ↓IL-6, ↓IKBKe | ↓Pro-inflammatory cytokines | 163 | |
MDA-MB-231 & ZR75.1 | ↓CXCL10, ↓IL-6, ↓IL-1α, ↓IRAK1, ↓IRAK4, ↓NF-κB, ↓p38-MAPK, ↓IP10 | ↓Impact of senescent cells on breast cancer cells, ↓fibroblast proliferation | 308 | |
BT-474 | ↓p-JAK1, ↓p-JAK2, ↓p-STAT3, ↓VEGF, ↑c-CASP-8, ↑c-CASP-3 | ↑Apoptosis, ↓proliferation, ↓colonization | 174 | |
MDA-MB-468 | ↓Akt | ↓Proliferation | 177 | |
MDA-MB-231 & T47D | ↑CASP-3, ↑c-PARP, ↑Bax, ↓Bcl-2, ↑LC3-II | ↑Apoptosis, ↓proliferation, ↓colonization, ↑autophagy | 309 | |
MCF-7/HER2 & MCF-7 vec | ↑c-CASP-8, ↓p-HER-2, ↑p53, ↑p21, ↓p-JAK1, ↓p-STAT3, ↓NF-κB, ↓p-IκBα | ↑Apoptosis, ↓proliferation | 175 | |
Silibinin | MCF-7 & T47D | ↓miR-21, ↓Bcl-2 | ↑Apoptosis | 149 |
MCF-7 | ↓Maspin, ↓ERα | ↓Proliferation | 310 | |
T47D | ↓hTERT, ↓cyclin D1 | ↓Proliferation | 147 | |
MCF-7 | ↓Bcl-2, ↑p53, ↑Bax, ↑BRCA1, ↑ATM | ↑Apoptosis, ↓proliferation | 145 | |
MCF-7 & T47D | ↑PTEN, ↑p21, ↓Bcl-2, ↑p27 | ↓Proliferation, ↑apoptosis, ↑necrosis | 311 | |
MCF-7 | ↓miR-21, ↓miR-155, ↑CASP-9, ↑BID | ↑Apoptosis, ↓proliferation | 148 | |
MCF-7 | ↓ERα, ↓Akt, ↓mTOR, ↓ERK, ↑CASP-6, ↑p53, ↓APAF-1, ↓p62, ↑Bax, ↓Bcl-2, ↑LC3-I to LC3-II | ↑Apoptosis, ↑autophagy | 151 | |
MCF-7 | ↑Atg12-Atg5, ↑LC3-1 to LC3II, ↑Beclin-1, ↓Bcl-2, ↑BNIP3, ↑ROS | ↑Autophagy | 150 | |
MCF-7 | ↑p53, ↑p21, ↑BRCA1, ↑Bak, ↑ATM, ↓Bcl-xl | ↑Apoptosis, ↓proliferation | 143 | |
MCF-7 & MDA-MB-231 | ↓ERK, ↓Akt, ↓Notch-1 | ↑Apoptosis | 312 | |
SKBR3 | ↓NF-κB | ↑Apoptosis, ↓proliferation | 142 | |
MDA-MB-468 | ↓EGFR, ↓VEGF, ↓COX-2, ↓MMP-9 | ↓Metastasis, ↓infiltration, ↓tumor volume | 144 | |
MCF-7 | ↓MMP-9, ↓MEK, ↓ERK | ↓Migration | 141 | |
MDA-MB-231 & T47D | ↓Wnt, ↓β-catenin, ↓LRP6 | ↓Proliferation | 313 | |
Pterostilbene | MDA-MB-468 | ↑ERK1/2, ↓cyclin D1, ↑p21, ↓Akt, ↓mTOR, ↑Bax | ↑Apoptosis, ↓proliferation | 189 |
MCF, MDA-MB-231 & Hs578t | ↑E-cadherin, ↓vimentin, ↓snail, ↓slug, ↓ZEB1, ↑miR-205, ↓Src/Fak | ↓Metastasis | 191 | |
MCF-7 | ↓CD44, ↑β-catenin, ↓hedgehog, ↓Akt, ↓GSK3β signalling, ↓cyclin D1, ↓c-Myc | ↓bCSCs, ↓mammospheres | 195 | |
MCF-7 & MDA-MB-231 | ↓NFκB, ↓vimentin, ↓Twist1, ↑E-cadherin | ↓bCSCs, ↓metastasis | 196 | |
MDA-MB-231 | ↓MMP-2, ↓MMP-9, ↓cortactin, ↓c-Src kinase, ↓MT1-MMP | ↓Metastasis | 314 | |
Sulforaphane | SUM-149 & SUM-159 | ↓NF-κB p65 subunit, ↓p52 | ↓bCSCs, ↓mammospheres, ↓proliferation | 204 |
MCF-7, MDA-MB-231 & SK-BR-3 | ↓DNMT1, ↓DNMT3B, ↑p21, ↑p27, ↓miR92b, ↓miR-23b, ↓miR-381, ↓miR-382, ↓Akt, ↓AMPK, ↓ATP | ↑Cellular senescence, ↑apoptosis, ↑autophagy | 210 | |
MCF-7 & MDA-MB-231 | ↓Akt, ↓NF-κB, ↓Bcl-2 | ↑Apoptosis, ↑sensitization to paclitaxel | 205 | |
MCF10DCIS.com | ↓TNF-α, ↓MMP-2, ↓MMP-9, ↓MMP-13 | ↓Migration, ↓invasion | 206 | |
MCF-7 | ↓Bcl-2, ↓COX-2 | ↑Apoptosis, ↓proliferation | 315 | |
MCF-7 | ↓MMP-9, ↓NF-κB | ↓Invasion | 207 | |
Luteolin | MDA-MB-231 (4175) LM2 | ↓VEGF | ↓Angiogenesis, ↓lung metastasis | 274 |
MCF-7 & MDA-MB-231 | ↓p-EGFR, ↓p-STAT3, ↓p-AKT, ↓p-ERK1/2 | ↓Proliferation | 316 | |
BT-474 & T47D | ↓VEGF, ↓CD44, ↓ALDH | ↓Angiogenesis, ↓proliferation, ↑apoptosis | 317 | |
MDA-MB-231, BT5-49 & female nude mice | ↓Vimentin, ↓slug, ↓β-catenin | ↓Lung metastasis | 275 | |
MDA-MB-231 | ↓Notch signalling, ↓VEGF, ↓cyclin D1, ↓MMP-2, ↓MMP-9, ↓Hes-1 | ↓Migration, ↓angiogenesis, ↓cell survival | 318 | |
MCF-7 (TAM-R) | ↓Cyclin E2 | ↓Tamoxifen resistance | 278 | |
MDA-MB-231 | ↓AKT, ↓cyclin A, ↓PLK1, ↓CDC2, ↓CDK2,↓ cyclin B1, ↓Bcl-xL, ↑p21, ↑Bax, ↓EGFR, ↓MAPK | ↑Apoptosis, ↓proliferation | 319 | |
MDA-MB-231, MCF-7 & SK-BR-3 | ↑ERK, ↑p38, ↑CASP-3, ↑c-PARP | ↑Apoptosis | 320 | |
MCF-7 | ↓IGF-1, ↓Akt, ↓ERα | ↑Apoptosis, ↓proliferation | 321 | |
Naringenin | MCF-7 (Tam-R) | ↓MAPK, CASP-7 | ↑Apoptosis, ↓proliferation | 322 |
4T1 | ↓TGF-β1, ↓PKC | ↓Pulmonary metastasis | 323 | |
MDA-MB-231 | ↓Integrin β3, ↓MMP-2, ↓MMP-9 | ↓Migration & invasion | 324 | |
HTB26 & HTB132 | ↓Bcl-2, ↓Cdk4, ↓Cdk6, Cdk7, ↓c-IAP-2, ↓x-IAP, ↑Bak, ↑Bax, ↑AIF, ↑CASP-3, ↑CASP-7, ↑CASP-8, ↑CASP-9, ↑p18, ↑p19, ↑p21, ↓pAkt, ↓P13K, ↓NFκB p65, ↓pIκBa | ↑Apoptosis, ↓proliferation, ↑chemo-sensitization | 325 | |
E0771 | ↑AMPK, ↓Bcl-2, ↓cyclin D1 | ↑Apoptosis, ↓proliferation | 326 | |
MDA-MB-231 | ↓Survivin, ↑p21, β-catenin | ↓Proliferation, ↑apoptosis | 339 | |
MCF-7 | ↓P13K, ↓MAPK, ↓ERK1/2, ↓AKT, ↑CASP-7, ↑CASP-9 | ↑Apoptosis, ↓proliferation | 328 | |
MCF-7, T47D & MDA-MB-231 | ↑CASP-3, ↓AKT, ↑p38 | ↑Apoptosis, ↓proliferation | 329 | |
α-Mangostin | T47D, MDA-MB-468, SKBR3 &AU565 | ↓Bcl-2, ↓Mcl-1, ↓P13K, ↓ERK1/2, ↓ERα, ↓HER2, ↓Akt, ↓ERK1/2, ↑p-p38, ↑p-JNK1/2, ↓MAPK | ↑Apoptosis, ↓proliferation, ↓colonization | 251 |
MCF-7 & MDA-MB-231 | ↑p53, ↑Bax, ↑PARP cleavage, ↓Bcl-2, ↓Bid, ↓pS2, ↓ERα, ↑CASP-7, ↑CASP-8, ↑CASP-9 | ↑Apoptosis, ↓proliferation | 254 | |
MCF-7 & MDA-MB-231 | ↓FASN, ↓FAK, ↓pAKT, ↑Bcl-2, ↓Bax, ↑p-ERK1/2 | ↑Apoptosis | 253 | |
MDA-MB-231 | ↑CASP-3 | ↑Apoptosis, ↓proliferation | 252 | |
MDA-MB-231 | ↑ p21cip1, ↑CASP-3, ↑CASP-8, ↑CASP-9, ↑CHEK2, ↓CDKs, ↓PCNA, ↓cdc(s) | ↑Apoptosis | 255 | |
Thymoquinone | MCF-7, T47D & EMT6/p | ↓VEGF, ↑IFN-γ, ↑IL-4 | ↓Angiogenesis, ↑apoptosis, ↓proliferation | 118 |
MCF-7 | ↓hsa04310 (Wnt), ↑hsa04115 (p53), ↓hsa04151 (P13K/AKT), ↓hsa04010 (MAPK) | ↑Apoptosis | 153 | |
MCF-7 | ↑p53 | ↑Apoptosis | 331 | |
EMT6/p | ↓VEGF, ↑IFN-γ, ↓IL-4, ↓AST, ↓ALT | ↑Apoptosis, ↓proliferation, ↑necrosis, ↓angiogenesis | 158 | |
MCF-7 | ↑PTPRR, ↓MAPK, ↓p38-MAPK, ↑TGF-β, ↓TP53, ↓Bcl-2, ↓CARD16, ↓EGF-EGFR, ↓GPCR | ↑Apoptosis | 152 | |
MCF-7 | ↑Bax, ↑p21, ↑Maspin, ↓Bcl-2, ↓HDAC | ↑Apoptosis, ↓proliferation, ↓migration | 155 | |
4T1, PMEF & Balb/c mice | ↑BRCA1, ↑p21, ↑HIC1, ↑CASP-3, ↑CASP-7, ↑CASP-12, ↑PARP, ↓p65, ↓p-Akt1 | ↑Apoptosis, ↓proliferation, ↓migration | 157 | |
MCF7 & MDA-MB-231 | ↑TGF-β, ↑E-cadherin, ↑cytokeratin 19, ↓MMP-2, ↓MMP-9, ↓integrin αV, ↓snail, ↓Twist, ↓Smad2, ↓NF-κB | ↑Apoptosis, ↓proliferation, ↓migration, ↓invasion, ↓colonization, ↑sensitization to radiation | 154 | |
MCF-7, T47D, MDA-MB-231 & MDA-MB-468 | ↓Cyclin D1, ↓cyclin E, ↓p27, ↓survivin, ↓Bcl-xl, ↓Bcl-2, ↑Bax, ↑PARP, ↑procaspase-3, ↑Cyt c, ↓Akt, ↑PTEN, ↓PDK1 | ↑Apoptosis, ↓proliferation, ↓viability | 332 | |
Isoliquiritigenin | MCF-7, MDA-MB-231, 4T1, BT474, MCF-10A | ↓miR-374a, ↑PTEN, ↓Akt, ↑Bax, ↓Bcl-2, ↑c-CASP-9, ↑Cyt C, ↑MMP-7, ↓p-GSK3β, ↓β-catenin | ↑Apoptosis, ↓proliferation, ↓lung metastasis, ↓migration, ↓invasion | 181 |
MDA-MB-231 & Hs-578T | ↓miR-21, ↓STAT3 | ↓Invasion | 184 | |
MDA-MB-231 & Hs-578T | ↑RECK, ↓miR-21 | ↓Invasion | 185 | |
MCF-7 & MDA-MB-231, MMTV-PyMT mice | ↓DNMT1, ↓β-catenin, ↑WIF1, ↓Wnt, ↓β-catenin, ↓Cyclin D1, ↓Survivin, ↓C-Myc, Oct-4 | ↓Proliferation, ↓lung metastasis, ↓initiation & progression, ↓bCSCs & its self-renewal | 187 | |
BT549 & MDA-MB-231 | ↓COX-2, ↓CYP 4A, ↓PGE2, ↓PLA2, ↑cleaved caspase-3 & −9 | ↓Metastasis, ↑apoptosis, ↑anoikis | 333 | |
BT549, MCF-7 & MDA-MB-231 | ↓β-catenin, ↓ABCG2, ↓GRP78, ↑proteasome degradation pathway, ↓CD44+CD24−/low, ↓Survivin, ↓Cyclin D1, ↓Oct-4, ↓c-Myc, ↓GSK-3β, | ↓Proliferation, ↓colonization, ↑apoptosis, ↓bCSCs self-renewal & differentiation, ↑chemosensitization | 188 | |
MCF-7, MCF-7/ADR & MCF-10A | ↓miR-25, ↓ABCG2, ↑ULK1, ↑LC3-II | ↓proliferation, ↑chemosensitization, ↓colonization | 186 | |
MDA-MB-231 | ↓VEGF, ↓HIF-1α, ↓MMP-2, ↓MMP-9, ↓p38, ↓Akt, ↓NF-κB, ↓P13K | ↓Migration, ↓proliferation, ↓angiogenesis | 48 | |
3,3′-Diindolylmethane | MCF-7 | ↓EMT, ↓CXCR4, ↓N-cadherin, ↑E-cadherin, ↓snail, ↓slug, ↓cathepsin B, ↓cathepsin D, ↓MMP-2, ↓MMP-9 | ↓Metastasis, ↓proliferation | 264 |
MCF-7 & T47D | ↑p21 | ↓Proliferation | 334 | |
MDA-MB-231 | ↓Akt | ↓Proliferation | 266 | |
Ginsenosides | MCF-7 | ↓CASP1, ↓INSL5, ↓OR52A1, ↑CLINT1, ↑ST3GAL4, ↑C1orf198 | ↓Proliferation, ↑apoptosis | 52 |
MDA-MB-453, MDA-MB-231 & BT-549 | ↓NF-κB, ↓Bcl-2, ↑Bax, ↑CASP-3 | ↑Chemosensitization, ↑apoptosis, ↓proliferation | 215 | |
T47D & BT-474 | ↑AMPK | ↓Proliferation | 218 | |
MCF-7 | ↓MMP-2, ↓MMP-9, ↓mTOR, ↓Akt, ↓p62, ↓JNK, ↓P13K, ↓VEGFA, ↓VEGFB, ↓VEGFC, ↓Beclin-1, ↑LC3-II | ↓Angiogenesis, ↓invasion, ↑autophagy | 217 | |
4T1 | ↓miR-18a, ↓Smad2 | ↓Metastasis | 335 | |
MCF-7 & MDA-MB-453 | ↑p21WAF1/cip1, ↑p53, ↑p15INK4B, ↓CDK4, ↓cyclin E2, ↓Cyclin D1, ↑CASP-6, ↑CASP-7, ↑CASP-8, ↑CASP-9, ↑p38 MAPKs, ↑Bax, ↑DR4, ↑DR5, ↓survivin, ↑PARP | ↓Proliferation, ↑apoptosis | 219 | |
MDA-MB-231 | ↓NF-κB, ↓Akt, ↓ERK | ↓Proliferation, ↑apoptosis | 216 | |
Benzyl isothiocyanate | MDA-MB-231, MCF-7, MDA-MB-468, T47D, HBL-100, Hs578t, BT474 | ↑ERK, ↑p53, ↑LKB1, ↑p73, ↓Ki-67, ↓survivin, ↓XIAP, ↑p-ERK, ↑CREB phosphorylation, ↓PRAS40 phosphorylation | ↓Proliferation, ↑apoptosis, ↓colonization | 39 |
SUM159, MCF-7 & MDA-MB-231 | ↑KLF4 | ↓bCSCs | 336 | |
SUM159, MDA-MB-231, MCF-7, & MDA-MB-361 | ↓BMI-1, ↓ALDH1, ↑Notch-4 | ↓bCSCs, ↑apoptosis, ↓migration | 162 | |
MDA-MB-231, MCF-7 & SUM159 | ↓Ron, ↓sfRon, ↑SOX-2, ↑Nanog, ↑Oct-4, ↓ALDH1 | ↓bCSCs, ↓mammospheres | 161 | |
MDA-MB-231, MCF-7 & SUM159 | ↑Notch2 | ↓Migration | 337 | |
MCF-7, MDA-MB-231, MDA-MB-468, MCF-10A | ↑FOXO1, ↓p62, ↓mTOR, ↑LC3-II | ↑Autophagy, ↓viability | 338 | |
MDA-MB-231 | ↓uPA, ↑PAI-1, ↓NF-κB, ↓p-Akt, ↓c-Met phosphorylation | ↓Migration & invasion | 160 | |
Secoisolariciresinol | MCF-7 | ↑Erk1/2, ↑PI3K/Akt expression | Modulate estradiol effect (possibly ↓cell proliferation) | 221 |
MCF-7 | ↓ERα and PR expression, ↑estradiol secretion | Modulate estradiol effect (possibly ↓cell proliferation) | 222 | |
MCF-7 | Opposite action of ENL and END on telomerase activity | Possible opposite effect on cell division | 223 | |
MDA-MB-231 | ↓uPA-induced plasmin activation ↓MMP-2 and MMP-9 (ECM-mediated remodelling) | ↓Proliferation, ↓migration, ↓metastasis | 225 | |
MDA-MB-231 | ↓Ki-67, PCNA & FoxM1 gene expression ↓cyclin A2, B1, B2 & E1 gene expression ↓phosphorylation of FAK/paxillin pathway | ↓Proliferation | 224 | |
SKBR3, MDA-MB-231 | ↑Cytotoxic effects of chemotherapeutic agents ↓FASN expression and activity | Improve efficiency of chemotherapeutic agents | 220 | |
MCF7, MDA-MB-231 | ↑Cytotoxic effects of chemotherapeutic agents ↓GSTP1 expression and activity | ↓Cell growth, ↑apoptosis improve efficiency of chemotherapeutic agents | 232 | |
Animal models (ACI rats) | ↓Ki-67 gene expression ↓SHBG & IGFBP-3 serum levels, ↑ESR2 (ERβ) | ↓Tumor progression and burden ↓cell proliferation & disphasia | 226 |
Fig. 6 Regulation of molecular targets by curcumin in breast cancer cells. Idea adopted from Liu and Chen (2013).396 |
Recently, curcumin was shown to induce apoptosis in MCF-7 cells through increased caspase-3 and caspase-9 expressions. Additionally, this compound suppressed miR-21 expression via up-regulation of PTEN/Akt signalling.66 Curcumin also reduced the proliferation of MCF-7 cells by decreasing nitric oxide and ROS levels. Synergistic anti-proliferative effect was observed when ECM proteins: fibronectin or collagen were used along with curcumin.67 Moreover, the compound upregulated Bax and downregulated Bcl-2 expressions in MDA-MB-231 cells, leading to growth inhibition and apoptosis induction.68 Fatty acid synthase inhibition by curcumin also resulted in breast cancer cells apoptosis.69 Curcumin downregulated flap endonuclease 1 (DNA repair enzyme) expression via Nrf2 stimulation thereby reduced the proliferation of MCF-7 cells.70
Similarly, curcumin inhibited the nuclear translocation of β-catenin, hence impeded the trans-activation of slug and subsequent restoration of E-cadherin which led to increased formation of E-cadherin/β-catenin complex and more β-catenin in cytosol to finally suppress bCSCs migration and EMT.71 Recently, hypomethylation reactivation of tumor suppressors by curcumin has also been presented. The compound reversed the hypermethylation status of Glutathione S-Transferase Pi 1 (GSTP1) gene in MCF-7 cells, leading to its reactivation.72 Likewise, it also activated Deleted in Liver Cancer 1 (DLC1) promoter by decreasing its methylation level via down-regulation of Sp1 transcription factor to constrain the expression of DNA methyltransferase 1 (DNMT1) in MDA-MB-361 cells.73 Curcumin was also shown to decrease the methylation status of tumor suppressor termed as Ras-association domain family protein 1A (RASSF1A), thereby activating it. This was achieved through interference of curcumin with DNMT1.74
Co-treatment of curcumin with other drugs/phytochemicals have greatly enhanced their therapeutic efficacy in various malignancies, including breast cancer. Recently, the combined effect of curcumin and metformin (oral anti-diabetic drug) was explored in breast cancer cell lines which markedly suppressed VEGF expression, activated Th2 immune response and induced Trp53-independent apoptosis.75 Similarly, arabinogalactan—a polysaccharide obtained from larch wood—in combination with curcumin induced in vivo and in vitro apoptosis in breast cancer cells by increasing reactive oxygen species (ROS), cleaved-caspase-3 and Bax/Bcl-2 ratio. Reduced glutathione, p53 overexpression and a change in mitochondrial membrane were also observed.76,77 Curcumin synergistically enhanced the effects of retinoic acid (RA) and IFN-β on breast cancer cells by upregulating GRIM-19 (cell death regulatory gene) via STAT3-dependent and- independent pathways.78 RA induction of apoptosis via CRABPII/RAR signalling pathway has been reported and curcumin was recently shown to upregulate CRABPII and RAR thus overcoming RA-resistance in TNBC cell lines.79 Similarly, curcumin sensitized MDR breast cancer cells to cisplatin via downregulation of CCAT1 and inactivation of P13K/Akt/mTOR pathway.80 Moreover, curcumin chemosensitized breast cancer cells to 5-fluorocuracil (drug which exerts its action by inhibiting thymidylate synthase (TS)) through TS-dependent downregulation of NF-κB.81
Curcumin inhibited the invasion and adhesion of MCF-7 cells by reducing the expression of uPA via activation of NF-κB.82 Furthermore, the compound suppressed lysophosphatidic acid-induced invasion and metastasis in MCF-7 cells by interfering with RhoA/ROCK/MMPs pathway.83 Similarly, inhibition of angiogenesis and tumor growth through downregulation of NF-κB and its related gene products like PECAM-1, p65 and cyclin D1 by curcumin in MDA-MB-231 cells has also been reported.84 Curcumin upregulated p16INK4A and several other tumor suppressors, inhibited JAK2 and STAT3 pathways, leading to decreased α-smooth muscle actin and invasion/migration abilities of breast cancer-associated fibroblasts. Curcumin also suppressed lamin B1 and triggered p16INK4A-dependent senescence in these fibroblasts.85 Matrix metalloproteinases (MMPs) possess a significant part in remodelling of the extracellular matrix. Tissue inhibitor of metalloproteinases (TIMPs) regulate the activities of MMPs. Curcumin regulated cell metastasis by up-regulating expression of TIMP1/4 genes and inhibiting MMP-2 and MMP-9 in breast cancer cells.86 Mo et al. (2012) revealed that curcumin inhibit invasion of MDA-MB-231 cells via reduction of p-ERK and Smad2 signalling.87 The compound also affected genes related to Epithelial–Mesenchymal Transition (EMT), leading to reduced expression of β-catenin, N-cadherin, E-cadherin, Twist1, slug, AXL, fibronectin and vimentin, subsequently inhibiting migration and invasion in breast cancer cells.88 Other pathways and genes modulated by curcumin are presented in Table 2.
EGCG induced apoptosis through downregulation of telomerase and P13K/AKT and increased Bax/Bcl-2 ratio and p53 expression in T47D cells. The expression of hTERT gene was decreased while that of CASP3, CASP9 and PTEN was increased.92 Huang et al. (2017) also observed an increase in p53 and decrease in Bcl-2 in MCF-7 cells.93 Moreover, EGCG suppressed the growth of MDA-MB-231 cells through downregulation of β-catenin, cyclin D1 and pAkt.94 In tumor microenvironment, tumor-associated macrophages (TAM) carry a significant role. EGCG reduced TAM infiltration and M2 macrophage polarization by upregulating miR-16, thereby decreased tumor growth in murine breast cancer model.95
EGCG suppressed heat shock protein 90 (Hsp90) and promoted the translocation of progesterone receptor B (PR-B) into the nucleus, consequently resulting in downregulation of ERα.96 Physiological concentrations of EGCG inhibited the growth of ERα-positive MCF-7 cells through reduction in ERα and insulin-like growth factor (IGF) binding protein 2 and increased p53 and p21 (tumour suppressors).97 Furthermore, the compound downregulated expression of ER-α36 and thus suppressed the growth of ER-negative breast cancer stem/progenitor cells.98
EGCG sensitized MCF-7 cells to 5-fluorouracil by regulating the expression of Bcl-xL.99 The compound also achieved sensitization to tamoxifen by reducing the expression of Nrf2 (transcription factor) in tamoxifen-resistant MCF-7 cells.100 Similarly, a reduction in MMP-2 and MMP-9 in doxorubicin resistant MCF-7 cells has also been observed.101
Fig. 7 Molecular targets regulated by genistein in cancer cells. Downward arrows (↓) represent decrease of activity/expression/secretion while upward arrows (↑) represent increase of activity/expression/secretion. Idea adopted from Varinska et al. (2015).397 |
Genistein resulted in reactivation of several tumor suppressor genes such as adenomatous polyposis coli (APC), mammary serpin peptidase inhibitor (SERPINB5), ataxia telangiectasia mutated (ATM) and PTEN in breast cancer cell lines by decreasing their methylation status. This was accomplished through reduced expression of DNMT1 by genistein.108 Moreover, it also suppressed cyclin D1 and DNMT-1 in MCF-7 cells, leading to BRCA1 CpG demethylation and consequent reactivation of BRCA1.40 Prepubertal exposure to Bisphenol A (BPA) changes the signalling pathways which may add to carcinogenesis, as revealed by DNA methylation studies. Conversely, prepubertal exposure to genistein or genistein + BPA resulted in hypomethylation of several genes in rat mammary tissues out of which HPSE and RPS9 genes were linked with enhanced long term survival, thus emphasizing cancer preventive properties of genistein.109 In another study, lifetime genistein intake improved response of mammary tumors to tamoxifen resistance in Sprague-Dawley rats and also reduced the risk of recurrence. Genistein administration downregulated the unfolded protein response (UPR) and genes linked to autophagy such as ATF4, GRP78, Beclin-1 and IRE1α, as well as, immunosuppression-linked genes like Foxp3 and TGFβ and upregulated CD8a (cytotoxic T-cell marker) in tumors. This demonstrates that the enhanced response to endocrine therapy was pre-programmed early in life.110
Avci et al. (2015) found that genistein suppress the growth of MCF-7 cells via upregulation of miR-23b.111 Furthermore, it inhibited NF-κB via Notch-1 signalling and reduced the growth of TNBC cells.111 Long-term low-dose genistein administration sensitized inflammatory breast cancer cell lines to radiation and reduced mammosphere formation and the growth of stem cell populations.112 Downregulation of Hedgehog–Gli1 signalling by genistein is also linked with reduced bCSCs.113
Fig. 8 Resveratrol-mediated regulation of molecular targets underlying breast cancer cells proliferation, apoptosis, EMT/metastasis, epigenetic responses and sensitization to chemotherapy. Downward arrows (↓) represent downregulation while upward arrows (↑) represent upregulation. Idea adopted from Sinha et al. (2016).398 |
Combination of RSV with other compounds greatly improved its efficacy and bioavailability. Co-treatment of thymoquinone and RSV increased apoptosis, induced geographic necrosis, reduced VEGF and increased serum levels of IFN-γ in breast cancer cell lines.118 Likewise, combination with salinomycin markedly enhanced the anti-cancer properties of RSV in breast cancer cells via downregulation of canonical Wnt signalling proteins and vimentin.119 Similarly, RSV and pterostilbene reactivated the expression of ERα in ERα-negative breast cancer cell lines by increasing active chromatin markers such as acetyl-H3lysine9 and acetyl-H3/4 in ERα promoter region. Reduction in the activity of DNMT and level of 5-methylcytosine along with alteration in histone acetyl transferase and HDAC were also noted.120 Enhanced apoptosis of MCF-7 cells occurred when Sorafenib (drug inhibitor of angiogenesis and tyrosine kinase) was used in combination with RSV. The treatment induced the expression of p53, enhanced intracellular ROS generation and Bax/Bcl-2 expressions while reduced mitochondrial membrane potential. Moreover, downregulation of cyclin B1 and cyclin D1 also occurred while cleaved caspase-3, -9, cleaved poly(ADP-ribose) polymerase (PARP) and apaf-1 were found to be upregulated.121 RSV and rapamycin prevented mTORC1 (mechanistic target of rapamycin), autophagy and Akt activation, consequently instigating apoptosis in breast cancer cells.122 Co-treatment with melatonin inhibited aromatase, thus making them suitable for hormonal treatment of breast cancer.123 RSV also sensitized resistant MCF-7/DOX cells to doxorubicin via downregulation of MDR1.124 Furthermore, the compound enhanced the cellular accumulation of doxorubicin by reducing ABC transporter genes, MRP1 and MDR1 in resistant breast cancer cells.54 In another study, down-regulation of HSP27 by RSV has been linked with sensitization to doxorubicin in resistant MCF-7 cells.125 Similarly, RSV chemosensitized resistant MCF-7 cells to tamoxifen by modulating Smad phosphorylation and endogenous TGF-β production,126 and to melphalan by arresting cell cycle at S phase.127
Resveratrol modulation of microRNAs has also been observed. Venkatadri et al. (2016) demonstrated that the compound modulate crucial tumor-suppressive microRNAs such as miR-122-5p, miR-125b-5p, miR-409-3p, miR-542-3p and miR-200c-3p in breast cancer cells. These microRNAs regulate key cell cycle and anti-apoptotic proteins like X-linked inhibitor of apoptosis protein, Bcl-2 and CDKs.128
Combination therapy of quercetin and ascorbic acid (vitamin C) along with therapeutic drugs such as paclitaxel or doxorubicin markedly enhanced their anti-cancer properties in breast cancer cells by significant reduction in S and Go/G1 phases.133 Quercetin downregulated Her-2 and upregulated ERα, thereby reversed tamoxifen resistance in MCF-7 cells.134 Pentagalloylglucose (5GG), which shows structural resemblance to (−)-EGCG, in combination with quercetin persuaded S-phase arrest and caused apoptosis in MDA-MB-231 cells via decreased expression of S-phase kinase, while G2/M-phase arrest and apoptosis in AU565 cells occurred through reduced expression of Her-2.135
Quercetin revealed anti-metastatic property in TNBC cells by modulating EMT markers. It increased E-cadherin and decreased vimentin, leading to mesenchymal-to-epithelial transition, associated with modulation of cyclin D1 and c-Myc (β-catenin target genes) and a change in β-catenin nuclear localization.136 The calcineurin/NFAT pathway play a key role in angiogenesis which was inhibited by quercetin in breast cancer xenograft via downregulation of VEGF, VEGFR2 and NFATc3.137 Quercetin up-regulated miR-146a and prevented proliferation in breast cancer cells. The compound also inhibited invasion via EGFR suppression and induced mitochondrial-mediated apoptosis.138
Co-treatment of silibinin with cisplatin or paclitaxel increased early apoptosis by decreasing Bcl-2 and increasing mRNA levels of Bax, ATM, BRCA1 and p53 in MCF-7 cells.145 Silibinin also sensitized resistant breast cancer cells to paclitaxel and doxorubicin through inhibition of the key oncogenic pathways encompassing ERK, AKT and STAT3 in doxorubicin-resistant MDA-MB-435 cells and paclitaxel-resistant MCF-7 cells at 400 μM concentration.146 Similarly, silibinin and chrysin synergistically inhibited proliferation of T47D cells via downregulation of cyclin D1 and hTERT.147
Silibinin reduced proliferation of MCF-7 cells via down-regulation of miR-155 and miR-21 and induced apoptosis through intrinsic and extrinsic pathways by upregulating their apoptotic targets such as BID and CASP-9.148 However, Jahanafrooz et al. (2017) presented that down-regulation of miR-21 upon silibinin treatment has minimal effect on its anti-tumorigenic property in breast cancer cells and that other pathways are responsible for its anti-apoptotic affect.149
Silibinin exerted autophagic cell death in MCF-7 cells via ROS-dependent mitochondrial dysfunction (ΔΨm) and a reduction in ATP levels involving BNIP3 (Bcl-2 interacting protein 3, a member of pro-death Bcl-2 protein family).150 However, according to Zheng et al. (2015), the anti-apoptotic and autophagy induction properties of silibinin in ERα-positive MCF-7 cells are due to down-regulation of ERα expression and subsequent inhibition of ERK and mTOR signalling pathways.151
TQ and tamoxifen synergistically induced apoptosis and decreased cell viability in both estrogen positive (MCF-7) and estrogen negative (MDA-MB-231) cell lines.156 The compound sensitized breast cancer cells to paclitaxel through multiple cascades involving extrinsic apoptosis, p53 signalling and tumor suppressor genes. Moreover, TQ regulated apoptosis inducible genes through death receptors and upregulated tumor suppressor genes including BRCA1, p21 and Hic1. Additionally, high doses of TQ downregulated pro-apoptotic factors like caspases and upregulated growth factors such as EGF and VEGF.157 Likewise, co-treatment of thymoquinone and melatonin greatly decreased tumor size via induction of apoptosis, activation of Th1 immune response and inhibition of angiogenesis.158
Truncated Recepteur d'Origine Nantais (sfRON) was also found as a novel mechanistic target for induction of apoptosis in breast cancer cells. BITC induced apoptosis in MDA-MB-361 and MCF-7 cells by regulating sfRON, whereby overexpression of sfRON in these cells intensified apoptosis, independent of JNK or MAPK hyperphosphorylation. Moreover, activation of Bak and Bax in sfRON overexpressing cells following BITC treatment were greatly enhanced while G2/M phase arrest and ROS generation were attenuated.159 However, according to Kim et al. (2013), full-length RON and sfRON overexpression offered protection against inhibition of bCSCs by BITC in MCF-7 cells and down-regulation of RON and its truncated form are associated with breast cancer stem cells inhibition by BITC.161 BITC inhibited bCSCs via downregulation of oncogene BMI-1 (Polycomb complex protein) and activation of Notch-4 in in vivo and in vitro breast cancer cells.162
Moreover, apigenin inhibited the proliferation and clonogenic survival of HER2-expressing BT-474 cells via caspase-dependent extrinsic apoptosis by upregulating cleaved caspase-3, cleaved caspase-8 and PARP cleavage. The compound decreased phospho-STAT3, phospho-JAK1 and phospho-JAK2 and prevented CoCl2-induced VEGF secretion and STAT3 nuclear translocation in HER2-expressing breast cancer cells.174 The same authors also mentioned before that apigenin induce apoptosis in HER2-overexpressing MCF-7 cells via extrinsic pathway, inhibit NF-κB and STAT3 and induce p53.175
Mechanism behind induction of cell cycle arrest by apigenin in MDA-MB-231 cells was also pointed out and it was shown that the compound suppress cyclin dependent kinase-1 (CDK1), cyclin A and cyclin B which are vital for G2-to-M-phase transition in cell cycle. Furthermore, apigenin increased p21WAF1/CIP1 and its interaction with proliferating cell nuclear antigen, thus obstructing cell cycle progression. Moreover, inhibition of histone deacetylase activity and an increase in acetylated histone H3 was also observed.176 Harrison et al. (2014) reported that apigenin reduce the proliferation of MDA-MB-468 cells by arresting cell cycle at G2/M phase and enhancing ROS production. Additionally, the compound reduced p-Akt.177
Peng et al. (2017) proposed that ISL induces apoptosis and prevent metastasis in breast cancer cells via downregulation of miR-374a. Decreased expression of miR-374a led to increased PTEN expression which prevented abnormal Akt signalling.181 ISL transcriptionally downregulated the expression of primary and mature miR-21 and decreased STAT3 signalling activity in breast cancer cells.184 The compound also suppressed breast cancer cells invasion by upregulating RECK (tumor suppressor gene) and downregulating miR-21.185 Moreover, ISL caused cell cycle arrest, chemosensitized and induced autophagy in MCF-7/ADR cells and also stimulated the degradation of ABCG2 through autophagy-lysosome pathway. Autophagy induction was associated with inhibition of miR-25 which led to increased expression of ULK1 (a kinase involved in autophagy).186 ISL treatment inhibited NF-κB, P13K/Akt and p38, ensuing decreased expressions of MMP-2, MMP-9, VEGF and HIF-1α which reduced the migration of breast cancer cells.48
ISL encouraged the demethylation of WIF1 promoter by docking into the catalytic domain of DNMT1, thereby increasing the gene expression of WIF1 and subsequently prevented mammary carcinogenesis by inhibiting bCSCs.187 The compound also chemosensitized bCSCs by inhibiting β-catenin/ABCG2 signalling via docking into ATP domain of GRP78 (binding immunoglobulin protein), resulting in inhibition of its ATPase activity and subsequent dissociation from β-catenin.188
Pterostilbene inhibited metastasis of TNBC cells through induction of miR-205 and negative modulation of EMT markers including vimentin, ZEB1, slug and snail and upregulation of E-cadherin. Downregulation of miR-205 caused decreased expression of Src—a nonreceptor tyrosine kinase whose activation and overexpression leads to different types of cancers, involving invasive breast cancer.191–194
Interestingly, pterostilbene and 6-shogaol inhibited bCSCs via reduced expression of CD44+ surface antigen and stimulated phosphorylation of β-catenin through hedgehog/Akt/GSK3β signalling inhibition thus downregulating downstream cyclin D1 and c-Myc.195 The occurrence of M2-polarized tumour associated macrophages (TAMs) enhanced metastatic abilities and CD44+/CD− CSCs in breast cancer cells. M2 TAMs-associated properties were suppressed by pterostilbene via inhibition of NF-κB, E-cadherin, vimentin and Twist1.196
Atwell et al. (2015) reviewed that most of the chemopreventive actions of SFN in breast and prostate cancers are due to modification of epigenetic mechanisms.208 Co-treatment of sulforaphane and withaferin A synergistically induced apoptosis and reduced cell viability and epigenetic processes in breast cancer cell lines through upregulation of Bax and downregulation of histone deacetylase 1 (HDAC1) and Bcl-2.209 On the other hand, SFN-induced senescence and cell cycle arrest in breast cancer cells are arbitrated by epigenetic alterations including decreased DNMT1 and DNMT3B expressions, global DNA hypo-methylation and variations in microRNA profile. The compound reduced methylation of N6-methyladenosine RNA (epigenetic regulation at RNA level) and exerted cytostatic action via induction of nitro-oxidative stress and downregulation of Akt signalling.210
Ginsenoside Rh2 provoked epigenetic methylation alteration in genes that are involved in immunity and tumorigenesis, thus increased immunogenicity and inhibited the growth of MCF-7 cells. Hyper-methylated genes like INSL5, OR52A1 and CASP1 experienced downregulation, while hypo-methylated genes including C1orf198, ST3GAL4 and CLINT1 displayed upregulation. Moreover, LINE1 (a global methylation marker) also displayed hypomethylation at specific CpGs.52 Rh2 also reversed resistance to docetaxel or adriamycin in resistant MCF-7 cells by differential microRNA expressions including miR-34a, miR-222 and miR-29a.214
Ginsenoside Rg3 enhanced cytotoxic effect of paclitaxel in TNBC cells via inhibition of NF-κB and Bcl-2 and upregulation of Bax and caspase-3.215 Kim et al. (2014) also reported that inhibition of NF-κB via inactivation of Akt and ERK is responsible for Rg3-induced apoptosis in breast cancer cells.216 Rg3 combined with recombinant human endostar suppressed the growth of breast cancer, inhibited cell invasion and angiogenesis and increased autophagy via decreased mRNA contents of MMP-2, MMP-9, VEGFA, VEGFB, VEGFC, p62, Beclin-1, P13K, mTOR, Akt and JNK.217
Similarly, ginsenoside Rg5 suppressed the proliferation of breast carcinoma via enhanced activation of AMPK and subsequent reduction in S6 and p70S6K activation.218 The compound induced apoptosis via regulation of Bax, cytochrome c and PARP and promoted cell cycle arrest at G0/G1 phase through downregulation of CDK4, cyclin E2 and cyclin D1 and upregulation of p21WAF1/CIP1, p53 and p15INK4B in breast cancer cell lines.219
Examination of the estrogenic activity of ENL using DNA microarray based gene expression profiling in MCF-7 cells revealed that both estradiol and ENL initiated the same estrogen signalling but their signals are then differentially and directionally modulated later in the pathway, resulting in the differences at cell function levels.221 Particularly, ENL have been shown to activate the ERK1/2 and PI3K/Akt pathways.221 ENL have significantly decreased both ERα and PR (progesterone receptor) expression and increased estradiol secretion in MCF-7 cells. Different dose–responses were observed between ENL and END, with END acting at highest concentration level.222 In the same way ENL and END produced opposite action on telomerase activity since ENL, but not END, have been shown to decrease telomerase activity in MCF-7 cells.223
Antiproliferative action of lignans was also observed in MDA-MB-231 breast cancer cells and has been attributed to the gene expression downregulation of Ki-67, PCNA and FoxM1.224 It also exerts a control on cell cycle by lowering gene expression of cyclin E1, A2, B1 and B2 and interfering with the cytoskeleton by downregulating phosphorylation of FAK/paxillin pathway, thus suppressing cell migration and invasion.224 ENL have been shown to suppress proliferation, migration and metastasis using MDA-MB-231 breast cancer cells through the reduction of uPA-induced plasmin activation and the matrix metalloproteases MMP-2 and MMP-9 mediated by ECM remodelling.225
A reduced tumor progression and burden was observed in a model deriving from ACI rats either with flaxseed or with SECO supplemented groups in both xerograph and carcinogen animal models of human breast cancer.226 Hypothesized mechanisms include reduction in estrogen related signalling, anti-angiogenic and antioxidant activity at higher doses. Decrease in serum levels of follicular phase SHBG and IGFBP-3 were noted in supplemented animals, whereas, ESR2 (ERβ) gene expression increased.226 Authors suggested that ESR2 can oppose or reverse the proliferation signalling in mammary epithelial tissue. SECO may activate this pathway, suggesting that increased ERβ signalling could be one mechanism behind the decrease of mammary proliferation and dysplasia observed with SECO administration. Together a decrease in the cell proliferation marker Ki-67 gene expression was also observed for flax and SECO supplemented animals. SECO also did not promote ovarian dysplasia in these models.226
High lignan exposure have been associated with reduced mortality in breast cancer patients. For examples recent studies have found high ENL blood concentrations or high dietary lignans intake to be associated with a better prognosis among post-menopausal women.227,228 In a population of 2182 breast cancer patients, ENL was found to be associated with reduced all-cause mortality and breast cancer-specific mortality. This association was restricted to early stage breast cancer and to patients with normal BMI.227 However, no association between ENL and HER2 status and Ki-67 gene expression were observed229 which is in good agreement with a smaller study (n = 24) that also reported no effect on Ki-67 gene expression together with no impact on both ERβ and CASPs expressions.230 On the contrary, a previously reported by Flower et al. (2005), a meta-analysis of 10 clinical studies evidenced both the antiproliferative and antiangiogenic activities for flax lignans.231 Interestingly, these effects appeared to be correlated with BMI, with significant actions restricted to normal BMI (<25), but without any significant correlation with the menopausal or the ER+/ER− status of the patients. These observational data suggested associations between flax intake and decreased risk of primary breast cancer, lower mortality among breast cancer patients and better mental health.231 A large pre-diagnostic plasma ENL levels and breast cancer prognosis among Danish post-menopausal women study evidence interesting results in crude models higher ENL levels were associated with lower risk of breast cancer-specific mortality but after adjustment for lifestyle factors this association was only borderline significant (P = 0.0501). However for women who never smoked or used hormones a more significant lower risk of mortality was noted and certainly deserve further investigations.229
Flaxseed lignan SECO and ENL were also reported to enhance the cytotoxic effect of classic chemotherapeutic agents (docetaxel, doxorubicin and carboplatin) in the metastatic breast cancer cell lines SKBR3 and MDA-MB-231, suggesting possible future direction in improving chemotherapeutic efficacy in breast cancer by adjuvant therapy with flaxseed lignans.220 Combination of dietary supplement along with the reduction in dosage of conventional breast cancer chemotherapeutic agent (doxorubicin) may retain its benefits while minimizing the cytotoxic side effects and thus may enhance its therapeutic efficiency.232 Interaction of SECO with gluthathione-S-transferase PI-1 (GSTP1) gene has also been observed. Inhibition of GSTP1 expression at both gene and protein level by SECO was confirmed in MCF-7 and MDA-MB-231 breast cancer cells. SECO was also shown to inhibit cell growth and increase cellular apoptosis by up-regulating pro-apoptotic Casp-3 and Bax expression and down-regulating anti-apoptotic Bcl-xl and Bcl-2 expression.232
Vinblastine is extensively used for the treatment of breast cancer and Kaposi's sarcoma.238,401 Vinblastine not only inhibits the tumor growth but also malignant angiogenesis and can specifically bind to tubulin, thereby restricting its polymerization and subsequent microtubules association.237 Recently, it was shown that vinblastine induced apoptosis is MCF-7 cells by interfering with microtubules. The compound increased PARP cleavage, however, it was demonstrated that colchicine was more effective than vinblastine in inducing apoptosis in docetaxel resistant breast cancer (MCF-7TXT) cells.238 Interestingly, it was observed that enhanced sensitization of MDA-MB-231 cells to vinblastine or actinomycin D can be achieved by disrupting the function of Golgi apparatus inside these cells, thus resulting in increased apoptosis and decreased cell migration and proliferation.239 Likewise, co-treatment of cells with vinblastine or actinomycin D and golgicide A or brefeldin A (disrupting agents of ADP-ribosylation factor 1 (ARF1)—a protein required for homeostasis of Golgi complex) caused the similar effects via reducing the levels of either phospho-AKT or phospho-ERK1/2.239 Furthermore, combined treatment of MDA-MB-231 cells with vinblastine and docetaxel decreased survivin expression and induced apoptosis. A synergistic effect was observed when deguelin (a survivin inhibitor) was used along with vinblastine and docetaxel, suggesting that survivin downregulation may be useful in increasing the therapeutic efficacy of these chemotherapeutics.240
Several obstacles limit the clinical efficacy of curcumin such as poor bioavailabilty, quick metabolism and slow aqueous solubility.345,346 To improve the potential clinical efficacy of curcumin in high-risk populations, present research is mainly focusing on enhancing bioavailability to overcome both rapid compound metabolism and variability of absorption. Several efforts combining glucuronidation inhibitors (piperine, for instance) with curcumin to restrict intestinal and hepatic metabolism have revealed promising results.347 Other recent attempts have focused on using nanoparticles, curcumin analogs or delivery via phospholipid or liposomal complexing.348,349 A phase-I dose-escalation trial revealed that participants getting single dose of liposomal curcumin had a dose-dependent enhancement in plasma level of curcumin, as well as, its active metabolite (tetrahydrocurcumin) without any clinical side effects. However, at higher doses of curcumin (120 mg m−2), changes in morphology of RBC were seen, signifying a dose restricting sign of toxicity.350
Despite the promising anti-proliferative effects of resveratrol in vitro and in animal models, its translation to clinics also face several challenges. One obstacle is limited bioavailability as resveratrol is metabolically excluded from the body very fast, thereby creating difficulty in maintaining a therapeutically effective level in bloodstream.351 Recently, several efforts have been made in combining other natural substances with resveratrol to enhance its overall therapeutic value, especially cancer prevention.351 Pharmacokinetic studies of resveratrol showed extensive and rapid metabolism to resveratrol-3-O-sulfate, resveratrol-3′-O-glucuronide and resveratrol-4′-O-glucuronide following oral administration at different doses, which doesn't allow for sufficient anticancer property of resveratrol.352,353 Thus, efforts are underway to somehow sustain its metabolism in order to achieve better tissue exposure in the body. As resveratrol is rapidly metabolized into its metabolites, therefore, it is unclear whether metabolites may have different biological activity than free resveratrol or not. Resveratrol-3-O-sulfate has also been found to be a chemopreventive agent.354 Another effort is to find the optimal route and dose of resveratrol.355
The main constituent of green tea i.e. EGCG, though extensively supported by results from cell culture, epidemiological, clinical and animal studies, however, there are several hurdles like bioavailability, stability and metabolic transformations under physiological circumstances.356 Several studies have displayed conflicting results concerning the cancer risk decreasing properties of green tea in different populations. This incompatibility in results may be because of variable tea preparations, variable bioavailability of compounds across populations and unknown concentration of antioxidants.340
Quercetin is also considered as a potent chemopreventive agent in various cancers, including breast cancer but its application in clinics is limited due to poor bioavailability, instability, low solubility and poor permeability.357 Several approaches have been developed to improve its bioavailability such as using nanoparticles, micelles, liposomes or inclusion complexes. Enhanced bioavailability will enable to bring this agent in forefront in disease therapeutics in the near future.357 Table 3 further depicts the bottlenecks involved in the clinical translation of various phytochemicals involved in breast cancer remedy.
Phytochemical | Bottlenecks in clinical translation | Ref. |
---|---|---|
Curcumin | Poor bioavailability, quick metabolism, slow aqueous solubility | 345 and 346 |
Resveratrol | Poor bioavailability, quick metabolism, lack of proper dose | 351 |
EGCG | Poor bioavailability, low stability, metabolic transformation under physiological circumstances | 356 |
Quercetin | Poor bioavailability, instability, low permeability, low solubility | 357 |
Apigenin | Poor bioavailability, poor aqueous solubility, low lipid solubility | 358 and 359 |
Kaempferol | Low bioavailability, poor solubility | 360 |
Genistein | Poor water solubility, low serum level after oral delivery (bioavailability), bitter taste | 361 and 362 |
Silibinin | Poor bioavailability, poor water and lipid solubility | 363 and 364 |
Parthenolide | Poor bioavailability & solubility in blood plasma, off-target effects, increased hydrophobicity | 258 |
Sulforaphane | Instability, poor gastrointestinal absorption, hydrophobicity, poor bioavailability | 365 and 366 |
3,3′-Diindolylmethane | Poor absoption, poor biodistribution | 367 |
Thymoquinone | Poor aqueous solubility, poor bioavailability, high lipophilicity | 368 and 369 |
Naringenin | Poor bioavailability, instability, low aqueous solubility, low permeability, extensive first pass metabolism | 370 and 371 |
Isoliquiritigenin | Poor bioavailability, poor solubility, low targeting ability, high effective dose | 187, 372 and 373 |
Ginsenosides | Poor oral absorption, poor bioavailability | 374 |
Secoisolariciresinol | Poor bioavailability | 375 |
Luteolin | Poor water solubility, poor bioavailability | 376 |
High costs and adverse reactions associated with the use of conventional drugs and irradiations reduced their therapeutic efficacy. In this regard, phytochemicals provide a safer and cost-effective approach. Most phytochemicals are readily available in vegetables, grains and fruits and their steady consumption would be effective to prevent breast cancer. It has been estimated that proper lifestyle adjustment could reduce cancer incidence by as much as two-thirds. Therefore, awareness should be raised among the general public about the proper and stable consumption of phytochemicals. Although, our understanding about multistage carcinogenesis has advanced, still the mechanism of action of most phytochemicals lacks adequate knowledge. The anticancer effects that most phytochemicals employ are probably the sum of several distinct mechanisms. Furthermore, deregulation or disruption of the intracellular signalling cascades sometime subject the cells to malignant transformation, so it is vital to recognize molecules in the signalling cascades that can be affected by specific phytochemicals in order to assess properly their underlying molecular mechanisms. Several phytochemicals have displayed non-toxic anti-tumor effects in xenograft model of nude mice, hence suggesting that they are effective in vivo.
A detailed insight about bioavailability and proper concentration of the required compounds are necessary, since most of them exerted dose-dependent inhibitory effects in breast cancer cells. Additionally, low concentration of some phytochemicals has shown to induce proliferation and activation of pro-survival autophagy in breast cancer, which further facilitate tumorigenesis. Likewise, large doses of phytochemicals resulted in development of additional diseases. For example, it has been shown that high doses of phytochemicals lead to chromosomal translocation, gastrointestinal disturbances and sexual dysfunction.377–379 Moreover, after absorption, phytochemicals are transformed to other conjugated forms, which may reduce their bioavailability. Thus, some alternations are required to enhance bioavailability of these phytochemicals, such as use of delivery systems like encapsulation, emulsion and other nanomedicine strategies. Pharmaceutical companies do not take interest in manufacturing phytochemical-based drugs due to intellectual property rights, which further creates hurdles. Some phytochemicals are produced by wild plants in relatively smaller proportions which make them difficult to be collected in large quantities, in which cases elicitation strategies and other plant tissue culture techniques provide a valuable insights for efficient accumulation of cancer related marker compounds, as well as, conservation of the respective plant species.
Strategies to decrease the side effects of chemotherapy are necessary, in which case, nanotechnology provides a useful opportunity in breast cancer therapy. The toxicity of conventional chemotherapeutic agents can be greatly reduced by administering them in nanoparticles and their combination with phytochemicals. Furthermore, new approach is required to enhance the intracellular stability, constant release and bioavailability of phytochemicals.380 Recently, it was reported that combination nanoparticles of paclitaxel and EGCG inhibited NF-κB activation, increased apoptosis, downregulated the genes involved in angiogenesis, cell survival and metastasis in MDA-MB-231 cell lines. Targeting the nanoparticles with anti-EGFR antibodies greatly enhanced these effects. Moreover, the EGCG-containing nanoparticles also overcame multidrug resistance in MDA-MB-231 cells via downregulation of P-glycoprotein.380 In another study, the intracellular concentration, stability and sustained release of EGCG was enhanced when loaded into chitosan-coated liposomes. The combined treatment significantly increased apoptosis and decreased cell viability.381 Similarly, quercetin encapsulation in MPEG-PLA (methoxypolyethylene glycol-polylactic acid) is useful in defeating its hydrophobicity. Quercetin nanoparticles induced apoptosis in MDA-MB-231 cells with sustained release for a duration of 10 days.382 In another report, quercetin-loaded mesoporous silica nanoparticles with folic acid tag ensured increased bioavailability and targeted delivery in breast cancer cells. The nanostructure caused apoptosis and cell cycle arrest in breast cancer cells via regulation of Bax and Akt signalling.383 Curcumin exerts numerous anticancer effects, however, it is faced with several limitations such as low bioavailability, quick metabolism, quick degradation and low aqueous solubility. Nanotechnology is revealing promising results in improving the bioavailability of hydrophobic agents such as curcumin. It was observed that curcumin-loaded PLGA-PEG greatly improved the cytotoxic effects in MCF-7 cell lines as compared to pure curcumin.384 Recently, curcumin-loaded folate-modified chitosan nanoparticles having targeted ability were developed which displayed good potential for breast cancer therapy. It was observed that reducing the pH of the release medium increased the release rate of curcumin from nanoparticles, showing pH responsive capacity of modified nanoparticles.385 Curcumin-loaded and calcium-doped dendritic mesoporous silica nanoparticles adapted with folic acid was also designed for effective treatment of breast cancer. This nanoformulation showed remarkable dispersal in aqueous solution, displayed pH responsive curcumin release and efficiently targeted the MCF-7 cells. The combination resulted in enhanced apoptosis, inhibition of proliferation, cell cycle arrest at G2/M phase, increased ROS generation and reduced mitochondrial membrane potential, in contrast to free curcumin.386 Resveratrol is also a promising candidate in breast cancer therapy, however, poor bioavailability and stability limit its clinical applications. Resveratrol-capped gold nanoparticles were synthesized previously, which repressed the TPA-induced invasion and migration abilities of breast cancer cells. The treatment greatly inhibited COX-2, ERK, MMP-9, NF-kB, P13K/Akt and AP-1.387 Resveratrol co-encapsulation with paclitaxel in a PEGylated liposome was successfully applied to drug resistant MCF-7/Adr cells which generated potent cytotoxicity. Moreover, the composite liposome also increased the bioavailability of the drug.388 In a recent study, resveratrol-load solid lipid nanoparticles were targeted on MDA-MB-231 cells which promoted Bax/Bcl-2 ratio and reduced the expression of c-Myc and cyclin D1, thereby greatly inhibiting proliferation, migration and invasion, as compared to free resveratrol.389 Thymoquinone-encapsulated nanoparticles were made by using hydrophilic and biodegradable polymers like PEG and PVP to overcome its poor aqueous solubility, minimal systemic bioavailability and light and thermal sensitivity. The resultant nanoparticles was more effective in killing cancer cells and less toxic to normal cells. Moreover, it displayed more potent anti-migratory activity on cancer cells.390 In another study, PLGA nanoparticles loaded with thymoquinone and paclitaxel were synthesized which showed improved anticancer activity in MCF-7 cells. This nanoformulation also aided in decreasing the toxic effects of paclitaxel by depressing its effective dosage.391 Similarly, nanostructured lipid carrier loading thymoquinone also improved the cytotoxicity and bioavailability of thymoquinone in MCF-7 and MDA-MB-231 cells.392 These results demonstrate that nanoformulations based on phytochemicals provide a useful strategy to enhance their cytotoxicity, bioavailability, stability and sustained release, as well as, to overcome drugs resistance and other harmful side effects associated with conventional chemotherapeutic agents.
This journal is © The Royal Society of Chemistry 2018 |