56
Views
9
CrossRef citations to date
0
Altmetric
Review

The Exosome And Breast Cancer Cell Plasticity

ORCID Icon & ORCID Icon
Pages 9817-9825 | Published online: 18 Nov 2019

Abstract

Cancer cell plasticity is the ability of cancer cells to reversibly interchange between distinct cell status, which plays a key role in cancer progression. Cancer cell plasticity is now known to be shaped by the secreted nanoparticles termed exosomes which transport proteins and lipids as well as nucleic acids. These aspects have emerged as key determinants of tumor progression and targeting, with approaches such as immunotherapy showing promise in the clinic. While significant strides have been made in this research area, some very interesting questions still warrant more and deeper investigation. We provide a review of the interplay between exosomes and breast cancer cell plasticity, and the potential implication in metastases and drug-resistance.

Introduction

Cancer cell plasticity refers to the ability to reversibly interchange between distinct cell status. It includes interconversion of different subtypes of cancer cell pools, activation of facultative cancer stem cells (CSCs), transdifferentiation or dedifferentiation, phenotypic transition of differentiated cells within a tumor to meet the challenges imposed by new microenvironments that accompany metastasis and by therapeutic interventions, and the dramatic habitat changes that accompany metastasis.Citation1 Breast cancer is the most commonly diagnosed cancer in women worldwide, also the second leading cause of cancer death among women after lung cancer, and accounts for more than 500,000 deaths annually worldwide.Citation2 It is also a complex heterogeneous disease which differs greatly among different patients (intertumoral heterogeneity) and even within each individual tumor (intratumor heterogeneity).Citation3 Breast CSCs have differentiation and transdifferentiation abilities. CSCs produce the original lineage cells similar to their normal stem cell counterparts. To promote tumor growth and metastasis in some tissue contexts, CSCs can also transdifferentiate into other lineage cells in addition to recruiting stromal cells from local or distant tissues. Following transformation and progression to malignancy, breast cancer cells do not remain inert but adapt to their systemic and local environment in order to evade death, proliferate and form metastases. This adaptive capacity is a property of cell plasticity. The forging of closer ties between preclinical, translational, and clinical research, together with advances in cancer models and single-cell technologies has revealed an unprecedented level of intra- and inter-tumoral heterogeneity and plasticity, and has started to reveal the pathway via which cancer cells circumvent therapeutic targeting. Cancer cell plasticity is also shaped by the secreted nanoparticles termed exosomes which can transport cellular contents such as proteins and lipids, as well as nucleic acids. Elements of contents of exosomes are now known to regulate cancer progression, tumor heterogeneity, and therapeutic resistance. While there have been significant strides in this research area, a lot of interesting questions still warrant deeper investigation. This review aimed to provide an in-depth viewpoint of the relations between the exosomes and breast cancer cell plasticity so as to better understand and defeat metastases and drug-resistance.

Plasticity In Histopathology Of Breast Cancer

Breast cancer varies in morphology, immunohistochemical profiles, and histopathological subtypes which have their unique clinical characteristics and individual outcomes. Morphologic plasticity in breast cancer is the representation of the histopathologic heterogeneity.Citation4 Breast cancer includes multiple histologic types, most are adenocarcinoma and invasive ductal cancer of no special type.Citation5,Citation6 WHO classification defined 21 distinct histological special types which include invasive lobular carcinoma, apocrine carcinoma, medullary carcinoma, adenoid cystic carcinoma, metaplastic carcinoma, micropapillary carcinoma, mucinous carcinoma, infiltrating ductal carcinoma with osteoclastic giant cells, neuroendocrine carcinoma, tubular carcinoma, invasive cribriform carcinoma, secretory carcinoma, lipid-rich carcinoma, glycogen-rich clear cell carcinoma, and so on. Different pathologic types have different prognosis and outcome following routine systemic therapy. Tubular, mucinous, medullary carcinoma and papillary carcinoma have favorable prognosis and better outcome than classic invasive ductal carcinoma.Citation6,Citation7 However, histological typing is not enough in clinical management decisions. Histological grade can provide complementary prognostic information which is based on the degree of differentiation, which also highlights the plasticity of breast cancer heterogeneity. The grade is divided into low, intermediate or high based on the morphological parameters, namely the percentage of the tumor arranged in glandular and tubular structures, the degree of nuclear pleomorphism, and the mitotic rate.Citation8 For decades, the histologic grade has been an important predictor of breast cancer outcome and helped to figure out what treatments might work best.Citation9,Citation10 In the last decade, gene expression profiling classified breast cancer into 5 intrinsic subtypes (Luminal A, Luminal B, Claudin-low, HER2-enriched, Basal-like) and a Normal Breast-like group.Citation11Citation13 Different subtypes of breast cancer differ in incidence, survival and response to treatment.Citation14Citation18 The molecular subtype information is complementary to classical clinical-pathological stage, and together can influence patients' outcome and response to the treatment.Citation19 For example, Luminal A subtype is associated with a low risk of local or regional recurrence,Citation1,Citation20 basal-like and triple-negative non-basal subtype have higher frequencies of relapse in lung, brain, and distant nodal metastasis than other subtypes.Citation21 Recent molecular research provides personalized treatment options which are based on significant numbers of publications in genomic profiling of breast cancer. Most molecular studies of breast cancer focuses on just one or two high information content platforms. Actually, breast cancer is a heterogeneous disease which is comprised of multiple distinct subtypes of cells that differ genetically, pathologically, and clinically. Plasticity also means “One tumor, different entities”.

The Biological Characteristics Of Exosome In Breast Cancer

An important “cross-talk” between cancer cells and its surrounding microenvironment is fundamental, just like “inter-sectional crosstalk of seed and soil”. Cells secrete extracellular vesicles (EVs) into their local environment or body fluids such as saliva, urine, serum, as well as cerebrospinal fluid, and so on. Exosomes are small EVs (30–100 nm in diameter) compared with microvesicles (50–1000 nm in diameter) and apoptotic bodies (1–5 μm in diameter).Citation22 The exosomes have pleiotropic functions in pathological and physiological processes, are novel mediators of cell-cell or cell-environment communication and activate signaling pathways in cells when they fuse or interact.Citation23,Citation24 Exosomes can fuse with multivesicular bodies through the plasma membrane after being secreted into the extracellular environment. Exosomes can be isolated by techniques such as ultracentrifugation, ultrafiltration and immunoprecipitation technologies which all exploit the characteristics of exosomes, such as their size, density, shape, and surface proteins, to aid their isolation.Citation25Citation27 The contents of exosomes may vary depending on cell of origin, status of activation and cell fate, but they have some particular contents, especially those involving vesicle biogenesis and intracellular sorting. They contain the proteins which are involved in membrane transport and fusion (flotillin, GTPases, annexins), tetraspanins family proteins (CD9, CD63, CD81 and CD82), heat-shock proteins (Hsp 60, Hsp70, Hsp90, HSPA5 and CCT2), proteins involved in biogenesis of multivesicular bodies such as TSG101 or ALIX, and lipid-bound proteins which account in part for the increased membrane rigidity relative to parent cell membranes.Citation28Citation35 Previous proteomic research indicated the exosomal proteome from MDA-MB-231 cells is distinct compared with MCF7. Periostin, integrin-β1, β-catenin, and N-Cadherin were enriched in the MDA-MB-231-derived exosomes compared with MCF7.Citation36 The tetraspanins family members (CD9, CD63, CD81 and Tetraspanin-14 antigens) are increased in the exosomes from MCF-7 compared with those from MDA-MB-231.Citation37

The Effect Of Exosome On The Dialog Between Breast Cancer Cells And Stromal Cells

The cancer surrounding stroma is the tumor-nourishing compartment in the tumor microenvironment responsible for the process of carcinogenesis and advancement. The stroma is composed of the extracellular matrix, endothelial cells, fibroblasts, adipocytes, and cells of the immune system which regulate the behavior of and co-evolve with tumor cells.Citation38,Citation39 The long-known “seed and soil” hypothesis for carcinogenesis and metastasis postulates that the appropriate host microenvironment (the soil) and the optimal growth of tumor cells are reciprocal.Citation40 The cancer cells and their microenvironment interact reciprocally as intimate partners during the progression of breast cancer.Citation41 Stromal cells provide matrix components or soluble factors that increase cancer cell survival and growth, which also promotes phenotypic plasticity in cancer cells, helping to acquire a more aggressive phenotype and influences treatment response.Citation42Citation44 Previous research indicated that RNA within exosomes transferred from stromal to breast cancer cells can activate STAT1-dependent antiviral signaling which is involved in the antiviral/NOTCH3 pathways in NOTCH signaling in breast cancer.Citation44 Both Notch pathway and antiviral/interferon signaling are known to regulate the maintenance of normal and cancer stem-like cells in cancer therapy resistance.Citation45 And breast cancer cells' exosome can destroy the tight junctions of vascular endothelial cells which are involved in the process of metastasis.Citation46 Cancer-associated fibroblasts (CAFs) are major stromal components which affect all aspects of tumor evolution, they build up and remodel the ECM structure through secretion of growth factors, cytokines, and chemokines.Citation47Citation49 The miRNA from the breast cancer exosome has been implicated in the intercellular crosstalk also. The breast-cancer-secreted, extracellular-vesicle-encapsulated miR-105 can mediate metabolic reprogramming of CAFs via MYC signaling. These CAFs in turn promote breast cancer growth by conditioning the shared metabolic environment. miR-105-reprogrammed CAFs promote glutamine and glucose metabolism to nourish adjacent breast cancer cells with sufficient nutrients, thus detoxifying metabolites under extreme metabolic conditions.Citation50 The exosomal G protein-coupled receptor, sphingosine-1-phosphate receptor 2 derived from MDA-MB-231, can promote CAFs' proliferation via activating ERK signaling.Citation51 The MMP-2 and MMP-9 in cancer cell exosomes can degrade components of the extracellular matrix and facilitate the aggressive cancer cells to invade surrounding tissue.Citation52

The Metabolism Plasticity And Exosome

An emerging hallmark of cancer is the altered metabolism, cancer cells experience complex metabolic rearrangement to sustain cancer growth by changes in metabolic pathways in biosynthetic processes and energy production.Citation53 The metabolic plasticity is paralleled by the metabolic interactions that occur between distinct tumor cell populations within the tumor, as well as between stoma and tumor.Citation54 Cancer cells of various origins displayed distinct metabolic strategies, and different tumor cell subtypes within a particular type of cancer can metabolically adapt due to distinct metabolic strategies.Citation55Citation59 The metabolic remodeling can satisfy the biosynthetic demand to support their abnormal proliferation and dissemination in nutrient-deprived and poorly oxygenated microenvironment.Citation60 Breast cancer metabolism heavily relies on aerobic glycolysis and glutamine catabolism to support cancer cell growth.Citation61Citation64 The different subtypes of breast cancer have different metabolisms. The triple-negative breast cancers (TNBC) typically related with the Warburg and mixed type, luminal type has obvious reverse Warburg and metabolic null type, estrogen receptor-positive breast cancers may rely on oxidative phosphorylation.Citation7,Citation55,Citation57Citation59,Citation65 And the hormonal therapy can abrogate oxidative phosphorylation generating self-renewal-deficient cancer cells in luminal breast cancer.Citation66 Notch signaling was enhanced to promote self-renewal of CSCs that display high glycolytic activity and aggressive hormone-independent tumor growth in vivo.Citation67Citation69 The Warburg type and the mixed type correlated with higher Ki-67 labeling indices which accompany high ATP synthase and glutaminase expression in stroma.Citation55 TNBC cells have special metabolic characteristics manifested by high glucose uptake, increased lactate production, and low mitochondrial respiration which is correlated with attenuation of mTOR pathway and decreased expression of p70S6K.Citation65 According to Warburg’s hypothesis, cancer cells are dominated by aerobic glycolysis as main mode of increased uptake of glutamine, glucose and aerobic glycolysis instead of more efficient oxidative phosphorylation. Cancer cells rely heavily on glucose and convert it to pyruvate through glycolysis rapidly. The glycolytic enzymes are commonly identified in the content of breast cancer exosome, such as enolase, aldolase, fructose bisphosphatase 1, triosephosphate isomerase, phosphoglycerate kinase, GADPH, and so on.Citation70,Citation71 They are also intrinsically associated with oncogenic switch, resistance to chemotherapy and radiotherapy.Citation72 Moreover, microRNA (miRNA) or long noncoding RNA (lncRNA) transferred by exosome is emerging as important regulators of cellular metabolism. The exosomal miR-155 of breast cancer cells triggers cancer-associated cachexia to promote metastasis on the catabolism of adipocytes and muscle cells via PPARγ. It promotes the beige/brown differentiation, remodeled resident adipocytes' metabolism through downregulating the expression of PPARγCitation73 Other research indicated that cancer-cell-secreted exosomal miR-122 can restrain glucose utilization through suppressing glycolytic enzyme pyruvate kinase in niche cells of pre-metastatic niches, which can reprogram energy metabolism to accommodate the massive energy needs of cancer cells during metastatic growth.Citation74 The exosomal HIF-1α-stabilizing lncRNA from tumor-associated macrophages inhibits glycolysis and apoptosis resistance of breast cancer cells.Citation75 The breast cancer cells can communicate through direct or indirect contact, such as the secretion of exosomes, to adapt to the shifting condition, metabolic cause lower glucose concentration and higher acidity subsequently suppressing infiltrated immune cells, contributing to cancer immune evasion and cancer aggressiveness.

Role Of Exosome In Plasticity In CSCs Of Breast

The hallmark feature of CSCs is reported to be self-renewal, and CSCs can differentiate into multiple subpopulations of cells within tumors.Citation76,Citation77 CSCs can regenerate tumors which recapitulate the heterogeneity of primary tumor from which they were isolated following orthotopic transplantation into mice. However, CSCs also induce resistance to anticancer therapy. The plasticity of the bidirectional conversion between non-CSCs to CSCs status is so complicated. The plasticity of CSCs refers to both reversible mesenchymal transitions and acquisition of stemness traits, which induce metastatic dissemination and development of resistance to treatments. The exosome derived from CSCs (CSCexo) contained selfrenewal promoting regulatory miRNAs, stemness specific proteins, and survival factors which can regulate tumor microenvironment and maintain tumor heterogeneity.Citation78 The CSCs reside in CSCs niches, which is a distinct protective microenvironment which regulates stemness, proliferation, and therapeutic resistance.Citation79,Citation80 The exosomal miRNAs of breast CSCs can promote the aggressiveness of cancer cells through nearby immune cells via interaction with toll-like receptors to up-regulate secretion of TNFα and IL-6 secretion.Citation81 And the dietary chemopreventive compound sulforaphane could promote exosomal miR-140 secretion of breast CSCs which prevents stemness in recipient cells in in vivo rat breast cancer models. The breast cancer stem cell exosome can modulate CSCs niche which is a vital aspect of exosome signaling in cancer. Lee et al revealed that exosome of osteogenic differentiating human adipose-derived stem cells can promote the drug resistance of breast CSCs by reprogramming of tumorigenic CSCs into non-tumorigenic cells, increasing the expression of osteogenic-related genes and decreasing the expression of drug-resistance genes such as ATP binding cassette transporter, the breast cancer gene family and the ErbB gene family.Citation82 Stemness-related molecules can be transferred from breast CSCs to non-CSCs by exosome, which leads non-CSCs to regain stemness phenotype. CSC‐exo induced dynamic or transient tumor plasticity in the tumor microenvironment.Citation83 It has also been investigated as potential therapeutic agents, so targeting the CSC-exosome transfer may have great potential for breast cancer therapy. The transcription factor ZEB1 and H3K27me3 histone modifications involved in the plasticity that the normal and CSC-like cells can arise de novo from more differentiated cell types and that hierarchical models of mammary stem cell biology should encompass bidirectional interconversions between stem and nonstem compartments.Citation84 The poised chromatin at ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity; the therapies targeting non-CSCs-to-CSCs plasticity should offer improved clinical outcome for breast cancer patients.Citation30,Citation85,Citation86 The interactions of CSCs and their surrounding microenvironment affect breast cancer cell malignancy directly and leads to tumor initiation, epithelial‐to‐mesenchymal transition (EMT), mesenchymal‐to‐epithelial transition (MET), metastasis, and therapeutic resistance. Recent single-cell studies in breast cancer have suggested that metastases derive from CSCs accompanied with increased MYC expression and CDK inhibition, which differentiate and undergo a switch from dormancy into proliferation as they colonize and produce more advanced metastatic tumors.Citation87 The stemness of hybrid epithelial/mesenchymal state in breast cancer is associated with poor survival; the plasticity to transition between EMT and MET can be the target to improve breast cancer patient survival independent of breast cancer-subtype.Citation88,Citation89 Due to most of current CSCs surface markers of breast appear to be present on embryonic or adult stem cells, and they are rarely expressed on normal breast tissue cells, e.g., CD10 and CXCR4.Citation90 In the future, multiple-antibody coated exosomes will need to be engineered to improve their CSCs targeting efficiency and to eradicate the CSCs and tumor plasticity, ideally.

Effects Of Exosome In EMT/MET Plasticity Of Breast Cancer Cells

EMT is a biologic process defined as the loss of epithelial characteristics and the acquisition of mesenchymal phenotype. Epithelial cells undergo multiple biochemical changes such as a loss of the epithelial traits of tight cell-cell adhesion and apico-basal polarization and a gain of invasiveness, enhanced migratory capacity, elevated resistance to apoptosis, and greatly increased production of ECM components. The reverse process of EMT is the transition from motile, multipolar or spindle-shaped mesenchymal cells to the epithelial cells, i.e., MET. The EMT/MET plasticity has been observed preclinically and clinically, whether any of these phenotypic transitions are indispensable for metastatic outgrowth remains unclear. It is involved in various pathophysiological processes including migration, treatment resistance and metastasis of breast cancer. Breast cancer cells acquire the increased motility and invasiveness along with EMT and re-epithelialize to form a metastatic solid mass under MET.Citation91,Citation92 Under EMT, cancer cells lose their polarity and cell-cell junctions and turn into a low proliferation state with increased migratory and invasion capabilities which are strongly associated with activation of Zeb (zinc finger and homeodomain proteins Zeb 1 and 2), Snail (zinc finger proteins Snail and Slug), and Twist (basic helix-loop-helix proteins E12, E47, Twist1, Twist 2 and Id) pathways.Citation93,Citation94 Once the cancer cells have reached the distant premetastatic niche, the reverse process takes place. It is a process called MErT (mesenchymal to epithelial reverting transition) which can return tumor cells to a high proliferative state and enables formation of macrometastases.Citation30,Citation86 The phenotypic plasticity that enables the crossover of EMT/MErT is necessary for tumor metastasis. EMT/MET plasticity implies switching on/off a set of genes which is mainly orchestrated by specific “master” transcription factors,Citation95,Citation96 miRNAs and lncRNAs. Twist1, ZEB1, ZEB2, Snail1 and Slug, as key EMT-inducing transcription factors, are involved in breast cancer metastasis through different signaling cascades such as serine/threonine-specific protein kinase (Akt), wingless-related integration site (Wnt), signal transducer and activator of transcription 3 (STAT3), and mitogen-activated protein kinase (MAPK) pathways, by repressing epithelial-related genes.Citation97Citation100 Exosomes contain active proteases capable of ECM degradation and remodeling by selectively and directly binding to the ECM-binding motif present on exosomal surface adhesion proteins.Citation101 Exosome biogenesis is enhanced by invadopodia and drives invasive behavior in cancer cells including breast cancer.Citation102 The exosomal miRNA secreted by breast cancer cells can enhance cell motility of normal fibroblasts and in turn is able to stimulate tumor cell migration by modulating its direct target, E-cadherin.Citation103 The normal hepatic niche-derived exosome can modulate MET process during seeding and suppression of tumor growth once the breast cancer cells have reached the liver.Citation104 Previous results indicated that the plasticity of EMT/MET phenotypes of breast cancer cells can be modulated by exosomes; primary cancer cells preserved their own niche and gave cells with aggressive traits necessary to colonize other free niches by exosome.Citation96 The communication resulted in relevant plasticity changes of gene expression of recipient cells in addition to microenvironment alterations. Exosome biogenesis is observed in immune cells, mesenchymal stem cells, neurons, fibroblasts, endothelial cells (ECs), and epithelial cells. Breast cancer cells secrete exosomes with specific capacity for cell-independent miRNA biogenesis, while normal cells lack this ability. Exosomes derived from cancer cells and serum from patients with breast cancer contain the RISC loading complex proteins, TRBP, Dicer and AGO2, which process precursor microRNAs into mature miRNAs.Citation105 The exosomes transferred from stromal to breast cancer cells can expand therapy-resistant breast cancer cells; RNA within exosomes stimulates the pattern recognition receptor RIG-I to activate STAT1-dependent antiviral signaling.Citation30

Plasticity-Based And Exosome Therapy For Breast Cancer

Breast cancer treatment includes surgery, chemotherapy, hormone therapy, radiation therapy, and targeted therapy. Plasticity in breast cancer cells within the same tumor is a reason for therapeutic resistance or later relapse because of genetic change, environmental differences, and reversible changes in cell properties. Some strategies target the tumorigenic cells as a result of minority populations of CSCs as they contribute to tumor growth and disease progression, while most other cancer cells have little or no capacity to drive tumor growth.Citation106,Citation107 A key question raised regarding the plasticity within the same tumor or among breast cancer patients is whether multiple pathways are important and whether they should be targeted simultaneously. Therapy failure may also contribute to tumor cell plasticity. The exosome has a close relationship with cancer cells' plasticity, so how can we make full use of plasticity-based and exosome therapy for breast cancer? First, CD47, HER-2, miR-21 and miR-1246 breast cancer patients' exosomal biomarkers, exosome-carrying TRPC5 and GSTP1 correlated with chemotherapy resistance, TRPC5, NEUROD1, HTR7, NANOG, HOXC and KISS1R in exosome were related with PFS, DFS or OS of breast cancer.Citation108 Second, the proposed targeting of the phenotypic plasticity will prove beneficial and to eradicate the exosome induced the key transcription factors involved in the alternation of EMT-MErT and non-CSCs-to-CSCs is providing new potential avenues for targeting the properties associated with cancer cell plasticity. Currently, post-translational modifications such as Ubiquitin and Ubiquitin-like modifiers in exosome were proposed to alter exosomal protein in cancer therapy.Citation109 Strategies to destroy the release of exosomes and exosome-mediated plasticity can potentially be exploited therapeutically in the future, including ESCRT (endosomal sorting complexes required for transport)-dependent and independent systems, tetraspanins and lipid-dependent mechanisms.Citation110 Third, as exosomes can mediate cell-to-cell communication, exosomes may be exploited as drug delivery vehicles with long-term safety and natural ability to carry intercellular nucleic acids and therapeutic molecules across membranes difficult to cross, such as BBB.Citation111 More research is needed.

Disclosure

The authors report no conflicts of interest in this work.

References

  • Varga J, Greten FR. Cell plasticity in epithelial homeostasis and tumorigenesis. Nat Cell Biol. 2017;19(10):1133–1141. doi:10.1038/ncb361128945230
  • DeSantis CE, Fedewa SA, Goding Sauer A, Kramer JL, Smith RA, Jemal A. Breast cancer statistics, 2015: convergence of incidence rates between black and white women. CA Cancer J Clin. 2016;66(1):31–42. doi:10.3322/caac.2132026513636
  • Turashvili G, Brogi E. Tumor heterogeneity in breast cancer. Front Med (Lausanne). 2017;4:227. doi:10.3389/fmed.2017.0022729276709
  • Gerashchenko TS, Zavyalova MV, Denisov EV, et al. Intratumoral morphological heterogeneity of breast cancer as an indicator of the metastatic potential and tumor chemosensitivity. Acta Naturae. 2017;9(1):56–67. doi:10.32607/20758251-2017-9-1-56-6728461975
  • Eheman CR, Shaw KM, Ryerson AB, Miller JW, Ajani UA, White MC. The changing incidence of in situ and invasive ductal and lobular breast carcinomas: United States, 1999–2004. Cancer Epidemiol Biomarkers Prev. 2009;18(6):1763–1769. doi:10.1158/1055-9965.EPI-08-108219454615
  • Makki J. Diversity of breast carcinoma: histological subtypes and clinical relevance. Clin Med Insights Pathol. 2015;8:23–31. doi:10.4137/CPath.S3156326740749
  • Ding S, Wu J, Lin C, et al. Predictors for survival and distribution of 21-gene recurrence score in patients with pure mucinous breast cancer: a SEER population-based retrospective analysis. Clin Breast Cancer. 2019;19(1):e66–e73. doi:10.1016/j.clbc.2018.10.00130396812
  • Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology. 1991;19(5):403–410. doi:10.1111/his.1991.19.issue-51757079
  • Neven P, Brouckaert O, Van Belle V, et al. In early-stage breast cancer, the estrogen receptor interacts with correlation between human epidermal growth factor receptor 2 status and age at diagnosis, tumor grade, and lymph node involvement. J Clin Oncol. 2008;26(10):1768–1769. author reply 1769–71. doi:10.1200/JCO.2007.15.614118519273
  • Rakha EA, El-Sayed ME, Lee AH, et al. Prognostic significance of Nottingham histologic grade in invasive breast carcinoma. J Clin Oncol. 2008;26(19):3153–3158. doi:10.1200/JCO.2007.15.598618490649
  • Perou CM, Sørlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752. doi:10.1038/3502109310963602
  • Sørlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98(19):10869–10874. doi:10.1073/pnas.19136709811553815
  • Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi:10.1038/nature1141223000897
  • Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295(21):2492–2502. doi:10.1001/jama.295.21.249216757721
  • Cheang MC, Chia SK, Voduc D, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst. 2009;101(10):736–750. doi:10.1093/jnci/djp08219436038
  • La Vecchia C, Bosetti C, Lucchini F, et al. Cancer mortality in Europe, 2000–2004, and an overview of trends since 1975. Ann Oncol. 2010;21(6):1323–1360. doi:10.1093/annonc/mdp53019948741
  • Millikan RC, Newman B, Tse CK, et al. Epidemiology of basal-like breast cancer. Breast Cancer Res Treat. 2008;109(1):123–139. doi:10.1007/s10549-007-9632-617578664
  • O’Brien KM, Cole SR, Tse CK, et al. Intrinsic breast tumor subtypes, race, and long-term survival in the Carolina Breast Cancer Study. Clin Cancer Res. 2010;16(24):6100–6110. doi:10.1158/1078-0432.CCR-10-153321169259
  • Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27(8):1160–1167. doi:10.1200/JCO.2008.18.137019204204
  • Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 2010;28(10):1684–1691. doi:10.1200/JCO.2009.24.928420194857
  • Kennecke H, Yerushalmi R, Woods R, et al. Metastatic behavior of breast cancer subtypes. J Clin Oncol. 2010;28(20):3271–3277. doi:10.1200/JCO.2009.25.982020498394
  • van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228.29339798
  • Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126(4):1208–1215. doi:10.1172/JCI8113527035812
  • Luga V, Zhang L, Viloria-Petit AM, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 2012;151(7):1542–1556. doi:10.1016/j.cell.2012.11.02423260141
  • Finkielsztein A, Mascarenhas L, Butin-Israeli V, Sumagin R. Isolation and characterization of neutrophil-derived microparticles for functional studies. J Vis Exp. 2018;(133).
  • Zarovni N, Corrado A, Guazzi P, et al. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods. 2015;87:46–58. doi:10.1016/j.ymeth.2015.05.02826044649
  • Zeringer E, Barta T, Li M, Vlassov AV. Strategies for isolation of exosomes. Cold Spring Harb Protoc. 2015;2015(4):319–323. doi:10.1101/pdb.top07447625834266
  • Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 2016;30(6):836–848. doi:10.1016/j.ccell.2016.10.00927960084
  • Madonna R, Van Laake LW, Davidson SM, et al. Position paper of the european society of cardiology working group cellular biology of the heart: cell-based therapies for myocardial repair and regeneration in ischemic heart disease and heart failure. Eur Heart J. 2016;37(23):1789–1798. doi:10.1093/eurheartj/ehw11327055812
  • Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14(3):195–208. doi:10.1038/nri362224566916
  • Latifkar A, Cerione RA, Antonyak MA. Probing the mechanisms of extracellular vesicle biogenesis and function in cancer. Biochem Soc Trans. 2018;46(5):1137–1146. doi:10.1042/BST2018052330301841
  • Record M, Silvente-Poirot S, Poirot M, Wakelam M. Extracellular vesicles: lipids as key components of their biogenesis and functions. J Lipid Res. 2018;59(8):1316–1324. doi:10.1194/jlr.E08617329764923
  • Maas S, Breakefield XO, Weaver AM. Extracellular Vesicles: unique Intercellular Delivery Vehicles. Trends Cell Biol. 2017;27(3):172–188. doi:10.1016/j.tcb.2016.11.00327979573
  • Hyenne V, Labouesse M, Goetz JG. The Small GTPase Ral orchestrates MVB biogenesis and exosome secretion. Small GTPases. 2018;9(6):445–451. doi:10.1080/21541248.2016.125137827875100
  • Conde-Vancells J, Rodriguez-Suarez E, Embade N, et al. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J Proteome Res. 2008;7(12):5157–5166. doi:10.1021/pr800488719367702
  • Vardaki I, Ceder S, Rutishauser D, Baltatzis G, Foukakis T, Panaretakis T. Periostin is identified as a putative metastatic marker in breast cancer-derived exosomes. Oncotarget. 2016;7(46):74966–74978. doi:10.18632/oncotarget.v7i4627589561
  • Harris DA, Patel SH, Gucek M, Hendrix A, Westbroek W, Taraska JW. Exosomes released from breast cancer carcinomas stimulate cell movement. PLoS One. 2015;10(3):e0117495. doi:10.1371/journal.pone.011749525798887
  • Kultti A, Li X, Jiang P, Thompson CB, Frost GI, Shepard HM. Therapeutic targeting of hyaluronan in the tumor stroma. Cancers (Basel). 2012;4(3):873–903. doi:10.3390/cancers403087324213471
  • Turley EA, Wood DK, McCarthy JB. Carcinoma cell hyaluronan as a “Portable” cancerized prometastatic microenvironment. Cancer Res. 2016;76(9):2507–2512. doi:10.1158/0008-5472.CAN-15-311427197262
  • Paget S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev. 1989;8(2):98–101.2673568
  • Ribelles N, Santonja A, Pajares B, Llácer C, Alba E. The seed and soil hypothesis revisited: current state of knowledge of inherited genes on prognosis in breast cancer. Cancer Treat Rev. 2014;40(2):293–299. doi:10.1016/j.ctrv.2013.09.01024112814
  • Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–598.27550820
  • Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19(11):1423–1437.24202395
  • Boelens MC, Wu TJ, Nabet BY, et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell. 2014;159(3):499–513. doi:10.1016/j.cell.2014.09.05125417103
  • Kux K, Pitsouli C. Tissue communication in regenerative inflammatory signaling: lessons from the fly gut. Front Cell Infect Microbiol. 2014;4:49. doi:10.3389/fcimb.2014.0004924795868
  • Yan W, Wu X, Zhou W, et al. Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat Cell Biol. 2018;20(5):597–609. doi:10.1038/s41556-018-0083-629662176
  • Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18(2):99–115. doi:10.1038/s41573-018-0004-130470818
  • Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392–1400. doi:10.1038/ncb165818037882
  • Goruppi S, Clocchiatti A, Dotto GP. A role for stromal autophagy in cancer-associated fibroblast activation. Autophagy. 2019;15(4):738–739. doi:10.1080/15548627.2019.156993630653445
  • Zhou W, Fong MY, Min Y, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25(4):501–515. doi:10.1016/j.ccr.2014.03.00724735924
  • El Buri A, Adams DR, Smith D, et al. The sphingosine 1-phosphate receptor 2 is shed in exosomes from breast cancer cells and is N-terminally processed to a short constitutively active form that promotes extracellular signal regulated kinase activation and DNA synthesis in fibroblasts. Oncotarget. 2018;9(50):29453–29467. doi:10.18632/oncotarget.2565830034630
  • Rilla K, Mustonen AM, Arasu UT, Härkönen K, Matilainen J, Nieminen P. Extracellular vesicles are integral and functional components of the extracellular matrix. Matrix Biol. 2019;75–76:201–219. doi:10.1016/j.matbio.2017.10.003
  • Hirschey MD, DeBerardinis RJ, Diehl A, et al. Dysregulated metabolism contributes to oncogenesis. Semin Cancer Biol. 2015;35(Suppl):S129–S150. doi:10.1016/j.semcancer.2015.10.00226454069
  • Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. 2015;34:111. doi:10.1186/s13046-015-0221-y26445347
  • Choi J, Kim DH, Jung WH, Koo JS. Metabolic interaction between cancer cells and stromal cells according to breast cancer molecular subtype. Breast Cancer Res. 2013;15(5):R78. doi:10.1186/bcr347224020991
  • Elia I, Schmieder R, Christen S, Fendt SM. Organ-specific cancer metabolism and its potential for therapy. Handb Exp Pharmacol. 2016;233:321–353.25912014
  • Hussien R, Brooks GA. Mitochondrial and plasma membrane lactate transporter and lactate dehydrogenase isoform expression in breast cancer cell lines. Physiol Genomics. 2011;43(5):255–264. doi:10.1152/physiolgenomics.00177.201021177384
  • Kung HN, Marks JR, Chi JT. Glutamine synthetase is a genetic determinant of cell type-specific glutamine independence in breast epithelia. PLoS Genet. 2011;7(8):e1002229.21852960
  • Timmerman LA, Holton T, Yuneva M, et al. Glutamine sensitivity analysis identifies the xCT antiporter as a common triple-negative breast tumor therapeutic target. Cancer Cell. 2013;24(4):450–465. doi:10.1016/j.ccr.2013.08.02024094812
  • Lazar I, Clement E, Attane C, Muller C, Nieto L. A new role for extracellular vesicles: how small vesicles can feed tumors’ big appetite. J Lipid Res. 2018;59(10):1793–1804. doi:10.1194/jlr.R08372529678957
  • Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–1033. doi:10.1126/science.116080919460998
  • Dang CV. Links between metabolism and cancer. Genes Dev. 2012;26(9):877–890. doi:10.1101/gad.189365.11222549953
  • Hensley CT, Wasti AT, DeBerardinis RJ. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J Clin Invest. 2013;123(9):3678–3684. doi:10.1172/JCI6960023999442
  • Sahar S, Sassone-Corsi P. Metabolism and cancer: the circadian clock connection. Nat Rev Cancer. 2009;9(12):886–896. doi:10.1038/nrc274719935677
  • Pelicano H, Zhang W, Liu J, et al. Mitochondrial dysfunction in some triple-negative breast cancer cell lines: role of mTOR pathway and therapeutic potential. Breast Cancer Res. 2014;16(5):434. doi:10.1186/s13058-014-0434-625209360
  • Sansone P, Ceccarelli C, Berishaj M, et al. Self-renewal of CD133(hi) cells by IL6/Notch3 signalling regulates endocrine resistance in metastatic breast cancer. Nat Commun. 2016;7:10442. doi:10.1038/ncomms1044226858125
  • Pannuti A, Foreman K, Rizzo P, et al. Targeting Notch to target cancer stem cells. Clin Cancer Res. 2010;16(12):3141–3152. doi:10.1158/1078-0432.CCR-09-282320530696
  • Landor SK, Mutvei AP, Mamaeva V, et al. Hypo- and hyperactivated Notch signaling induce a glycolytic switch through distinct mechanisms. Proc Natl Acad Sci U S A. 2011;108(46):18814–18819. doi:10.1073/pnas.110494310822065781
  • Rizzo P, Miao H, D’Souza G, et al. Cross-talk between notch and the estrogen receptor in breast cancer suggests novel therapeutic approaches. Cancer Res. 2008;68(13):5226–5235. doi:10.1158/0008-5472.CAN-07-574418593923
  • Griffiths SG, Cormier MT, Clayton A, Doucette AA. Differential proteome analysis of extracellular vesicles from breast cancer cell lines by chaperone affinity enrichment. Proteomes. 2017;5(4). doi:10.3390/proteomes5040025
  • Ghosh A, Davey M, Chute IC, et al. Rapid isolation of extracellular vesicles from cell culture and biological fluids using a synthetic peptide with specific affinity for heat shock proteins. PLoS One. 2014;9(10):e110443. doi:10.1371/journal.pone.011044325329303
  • Muñoz-Pinedo C, El Mjiyad N, Ricci JE. Cancer metabolism: current perspectives and future directions. Cell Death Dis. 2012;3:e248. doi:10.1038/cddis.2011.12322237205
  • Wu Q, Sun S, Li Z, et al. Breast cancer-released exosomes trigger cancer-associated cachexia to promote tumor progression. Adipocyte. 2019;8(1):31–45. doi:10.1080/21623945.2018.155168830474469
  • Fong MY, Zhou W, Liu L, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17(2):183–194. doi:10.1038/ncb309425621950
  • Chen F, Chen J, Yang L, et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol. 2019;21(4):498–510. doi:10.1038/s41556-019-0299-030936474
  • Frank NY, Schatton T, Frank MH. The therapeutic promise of the cancer stem cell concept. J Clin Invest. 2010;120(1):41–50. doi:10.1172/JCI4100420051635
  • Li Y, Welm B, Podsypanina K, et al. Evidence that transgenes encoding components of the Wnt signaling pathway preferentially induce mammary cancers from progenitor cells. Proc Natl Acad Sci U S A. 2003;100(26):15853–15858. doi:10.1073/pnas.213682510014668450
  • Sharma A. Role of stem cell derived exosomes in tumor biology. Int J Cancer. 2018;142(6):1086–1092. doi:10.1002/ijc.v142.628983919
  • Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells. Cell Stem Cell. 2015;16(3):225–238. doi:10.1016/j.stem.2015.02.01525748930
  • Borovski T. De Sousa E, Melo F, Vermeulen L, Medema JP. Cancer Stem Cell Niche: the Place to Be. Cancer Res. 2011;71(3):634–639.21266356
  • Li Q, Eades G, Yao Y, Zhang Y, Zhou Q. Characterization of a stem-like subpopulation in basal-like ductal carcinoma in situ (DCIS) lesions. J Biol Chem. 2014;289(3):1303–1312. doi:10.1074/jbc.M113.50227824297178
  • Lee KS, Choi JS, Cho YW. Reprogramming of cancer stem cells into non-tumorigenic cells using stem cell exosomes for cancer therapy. Biochem Biophys Res Commun. 2019;512(3):511–516. doi:10.1016/j.bbrc.2019.03.07230905410
  • Fatima F, Nawaz M. Stem cell-derived exosomes: roles in stromal remodeling, tumor progression, and cancer immunotherapy. Chin J Cancer. 2015;34(12):541–553. doi:10.1186/s40880-015-0051-526369565
  • Chaffer CL, Brueckmann I, Scheel C, et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci U S A. 2011;108(19):7950–7955. doi:10.1073/pnas.110245410821498687
  • O’Flanagan CH, Rossi EL, McDonell SB, et al. Metabolic reprogramming underlies metastatic potential in an obesity-responsive murine model of metastatic triple negative breast cancer. NPJ Breast Cancer. 2017;3:26. doi:10.1038/s41523-017-0027-528748213
  • Subramanian A, Gupta V, Sarkar S, et al. Exosomes in carcinogenesis: molecular palkis carry signals for the regulation of cancer progression and metastasis. J Cell Commun Signal. 2016;10(3):241–249. doi:10.1007/s12079-016-0338-627473382
  • Lawson DA, Bhakta NR, Kessenbrock K, et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature. 2015;526(7571):131–135. doi:10.1038/nature1526026416748
  • Grosse-Wilde A, Fouquier d’Hérouël A, McIntosh E, et al. Stemness of the hybrid epithelial/mesenchymal state in breast cancer and its association with poor survival. PLoS One. 2015;10(5):e0126522. doi:10.1371/journal.pone.012652226020648
  • Luo M, Brooks M, Wicha MS. Epithelial-mesenchymal plasticity of breast cancer stem cells: implications for metastasis and therapeutic resistance. Curr Pharm Des. 2015;21(10):1301–1310. doi:10.2174/138161282166614121112060425506895
  • Kim WT, Ryu CJ. Cancer stem cell surface markers on normal stem cells. BMB Rep. 2017;50(6):285–298. doi:10.5483/BMBRep.2017.50.6.03928270302
  • Bigagli E, Cinci L, D’Ambrosio M, Luceri C. Transcriptomic characterization, chemosensitivity and regulatory effects of exosomes in spontaneous EMT/MET transitions of breast cancer cells. Cancer Genomics Proteomics. 2019;16(3):163–173. doi:10.21873/cgp.2012231018947
  • Pang YP, Vummenthala A, Mishra RK, et al. Potent new small-molecule inhibitor of botulinum neurotoxin serotype A endopeptidase developed by synthesis-based computer-aided molecular design. PLoS One. 2009;4(11):e7730. doi:10.1371/journal.pone.000773019901994
  • Hayasaki Y, Zhou C, Popescu G, Onural L. Feature issue of digital holography and 3D imaging (DH) introduction. Opt Express. 2014;22(23):29117–29118. doi:10.1364/OE.22.02911725402150
  • Kotiyal S, Bhattacharya S. Breast cancer stem cells, EMT and therapeutic targets. Biochem Biophys Res Commun. 2014;453(1):112–116. doi:10.1016/j.bbrc.2014.09.06925261721
  • Battistelli C, Cicchini C, Santangelo L, et al. The snail repressor recruits EZH2 to specific genomic sites through the enrollment of the lncRNA HOTAIR in epithelial-to-mesenchymal transition. Oncogene. 2017;36(7):942–955. doi:10.1038/onc.2016.26027452518
  • Aigner K, Dampier B, Descovich L, et al. The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene. 2007;26(49):6979–6988. doi:10.1038/sj.onc.121050817486063
  • Xu Y, Lee DK, Feng Z, et al. Breast tumor cell-specific knockout of twist1 inhibits cancer cell plasticity, dissemination, and lung metastasis in mice. Proc Natl Acad Sci U S A. 2017;114(43):11494–11499. doi:10.1073/pnas.161809111429073077
  • Wan T, Zhang T, Si X, Zhou Y. Overexpression of EMT-inducing transcription factors as a potential poor prognostic factor for hepatocellular carcinoma in Asian populations: a meta-analysis. Oncotarget. 2017;8(35):59500–59508. doi:10.18632/oncotarget.v8i3528938653
  • Imani S, Hosseinifard H, Cheng J, Wei C, Fu J. Prognostic value of EMT-inducing transcription factors (EMT-TFs) in metastatic breast cancer: a systematic review and meta-analysis. Sci Rep. 2016;6:28587. doi:10.1038/srep2858727335258
  • Lehmann W, Mossmann D, Kleemann J, et al. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat Commun. 2016;7:10498. doi:10.1038/ncomms1049826876920
  • Mu W, Rana S, Zöller M. Host matrix modulation by tumor exosomes promotes motility and invasiveness. Neoplasia. 2013;15(8):875–887. doi:10.1593/neo.1378623908589
  • Hoshino D, Kirkbride KC, Costello K, et al. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013;5(5):1159–1168. doi:10.1016/j.celrep.2013.10.05024290760
  • Baroni S, Romero-Cordoba S, Plantamura I, et al. Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 2016;7(7):e2312. doi:10.1038/cddis.2016.22427468688
  • Dioufa N, Clark AM, Ma B, Beckwitt CH, Wells A. Bi-directional exosome-driven intercommunication between the hepatic niche and cancer cells. Mol Cancer. 2017;16(1):172. doi:10.1186/s12943-017-0740-629137633
  • Melo SA, Sugimoto H, O’Connell JT, et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–721. doi:10.1016/j.ccell.2014.09.00525446899
  • Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–3988. doi:10.1073/pnas.053029110012629218
  • Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501(7467):328–337. doi:10.1038/nature1262424048065
  • Wang M, Ji S, Shao G, et al. Effect of exosome biomarkers for diagnosis and prognosis of breast cancer patients. Clin Transl Oncol. 2018;20(7):906–911. doi:10.1007/s12094-017-1805-029143228
  • Moreno-Gonzalo O, Fernandez-Delgado I, Sanchez-Madrid F. Post-translational add-ons mark the path in exosomal protein sorting. Cell Mol Life Sci. 2018;75(1):1–19. doi:10.1007/s00018-017-2690-y29080091
  • Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193–208.28733901
  • Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin B. 2016;6(4):287–296. doi:10.1016/j.apsb.2016.02.00127471669