Keywords
Extracellular matrix, breast cancer, metastasis, matrix metalloproteinases
Extracellular matrix, breast cancer, metastasis, matrix metalloproteinases
This version includes more details of role of ECM in breast cancer development and metastasis. The EMT process has been described with more stress on pathways involved in this process. A new subheading has been included to discuss the TME as therapeutic target. All the queries of referees have been answered in this version.
See the authors' detailed response to the review by Ren Xu
See the authors' detailed response to the review by Yunus A. Luqmani
See the authors' detailed response to the review by Andrew R. Craig
Breast cancer (BC) accounts for 25% of all cancer cases in women, and 12% of overall cancer cases worldwide1. The extracellular matrix (ECM) plays a crucial role in BC progression, invasion, and metastasis; thus, elucidating the role of ECM will help in designing therapies targeting different ECM components. Comprehensive studies are currently going on related to the involvement of ECM in BC progression, and this review focuses on the latest developments in this regard with possible molecular targets for therapies.
The ECM (includes basement membrane (BM) and stroma) interacts with the cells mediated by ECM receptors like integrins, discoidin domain receptor, syndecans, CD44, dystoglycans, and Rhamm2,3. The BM is mainly composed of laminins (laminin- 111 is involved in milk protein synthesis and secretion), type IV collagen, entactin, and proteoglycans. The stromal cells, adipocytes, and immune cells produce many ECM proteins like type I, II, and III fibrillar collagens, fibronectin, vitronectin, elastin etc. and the stroma is highly charged and hydrated, providing tensile strength to tissues4. It is observed fibrillar collagen I guides epithelial cell branching during the mammary gland development and macrophages are involved in this process of long fibre organization, required for branching morphogenesis (Rac1 acts as a modulator of collagen 1 orientation)5,6. Deregulation of the ECM dynamics is a hallmark of cancer. The ECM remodeling enzymes are deregulated changing the basic properties of ECM7. There is more deposition of collagens (COL I, II, III, V, IX), and overproduction of ECM components like heparin sulphate proteoglycans and CD44 which promote growth factor signaling in cancerous cells8,9. The ECM of cancerous tissue differs from that of normal tissue in following manners: the stroma of cancerous tissue seems to be stiffer than that of normal one; the COL I fibrils in BC tissue are highly linearized, and properly oriented, whereas relaxed nonoriented fibrils are observed in normal breast tissue; many MMPs are overproduced in cancerous tissues7,10–12. In the BC development the collagen I stiffens the ECM, thus promoting the tumor invasion and metastasis; whereas the BM prevents invasion by acting as a barrier3,13,14.
The EMT (process of losing epithelial characteristics and gaining mesenchymal properties) plays a significant role in the progression of tumor and metastasis, involving different transcription factors (TFs) and signals15–18. This process is characterized by loss of E-cadherin (cell-cell adhesion molecule) and cytokeratins, along with gain of N-cadherin, fibronectin, and vimentin (mesenchymal cell associated proteins) and this is termed cadherin switching (E – cadherin to N – cadherin)19. The EMT is regulated by different signaling pathways such as TGF-β, notch, and wnt pathway. All these pathways converge to activate the EMT – specific TFs such as Snail (SNAI 1), slug (SNAI 2), Zeb, and Twist which differentially express in cancerous cells to promote EMT20–22. Snail is a transcriptional repressor of E-cadherin (cell-cell adhesion molecule), and E-cadherin loss is a hallmark of EMT2. Snail and TWIST cooperate inducing another TF, ZEB123 (significant inducer of EMT, invasion, and metastasis), which is triggered by extracellular hyaluronic acid (HA). Furthermore, ZEB1 induces HAS2 synthesis, promoting HA production in a positive feedback loop and its expression is correlated with ZEB1 expression in poor prognosis tumors. HAS2 also has a role in TGF-β-induced EMT24. Platelets and platelet-derived TGF-β promote epithelial-mesenchymal-like transition and promote metastasis in vivo25. Both the canonical (Smad dependent pathway) and noncanonical (Smad independent pathway) TGF-β signaling activate the TFs (Snail, Zeb, Twist, and Six1) responsible for EMT. The TGF-β induced EMT might be facilitated through enhanced expression of PARP3 (Poly ADP-Ribose Polymerase 3) protein which promotes cell motility, and chemoresistance in breast epithelial cells26. The PARP3 seems to promote stemness of cancerous cells by inducing stem cell markers SOX2 and OCT4, and increasing the population of CD44high/CD24low tumor initiating cells. The notch signaling induces the TFs (Snail, Slug, Twist, and Zeb1/Zeb2) by acting through NF-κB, promoting cytokine production and cell survival. The wnt pathway induces Snail, thus down regulates E-cadherin via β-catenin. Besides these signaling, the hypoxic microenvironment also changes the function of mitochondria leading to HIF1 stimulation and subsequently increased expression of Zeb1 required for EMT22. The inflammatory factors such as TNF-α and IL-1β showed to induce plasticity in nontransformed breast epithelial cells (surrounding the transformed tumor cells) by initiating EMT through Snail and Zeb127. Apart from this, the steroid nuclear receptors such as estrogen receptors, progesterone receptors, glucocorticoid receptors, and mineralocorticoid receptors are also observed to regulate the expression of TFs inducing EMT28. A calcium - dependent phospholipid binding protein such as annexin A2 likely promotes EMT through activation of EGF/EGFR pathway. It is also observed annexin A2 directly binds to STAT3, which is a key EMT inducer up regulating the expression of TFs for EMT29. Aberrant cancer metabolism promotes EMT which further aggravates metabolism (especially glucose metabolism) through Snail and Twist30. One of the TF Runx1 (highly expressed in epithelium) is found to stabilize the mammary epithelial cell (MEC) phenotype thus prevents the EMT31. It is observed EMT is activated by ECM stiffness, which induces the release of TWIST1 from its anchor G3BP2 and this TF enters the nucleus and transcriptionally boost the EMT process through integrin clustering and activation32.
The cancer cells need to overcome anoikis (apoptosis due to loss of attachment to ECM) for metastasis event as this is a crucial barrier preventing tumor cell migration to secondary sites. Induction of anoikis occurs through lysosome – mediated down regulation of epidermal growth factor receptors (EGFRs) resulting in the termination of prosurvival signaling. It is observed the depletion of one of the kinases pre-mRNA splicing factor 4 kinase (PRP4K) promotes increased resistance to anoikis through reduced EGFR degradation (after cell detachment from ECM) with increased level of TrkB, vimentin, and ZEB133. Anoikis is evaded in ErbB2 expressing cells by multicellular aggregation during ECM detachment. EGFRs are stabilized by this aggregation, which results in ERK/MAPK survival signaling34. EGFRs could be the therapeutic targets to eliminate the ECM-detached cancer cells.
Various ECM-remodeling enzymes are induced in BC promoting stem/progenitor signaling pathways and metastasis. The different pathways that are regulated by ECM during remodeling in BC development, are wnt, PI3K/AKT, ERK, JNK, Src-FAK etc3,11,35. Major ECM proteins induced are fibrillar collagens, fibronectin, specific laminins, proteoglycans, and matricellular proteins and these could be potential drug targets for therapy36. Matrix metalloproteinases (MMPs) degrade ECM proteins promoting invasion and metastasis. The MMP-11 (stromelysin-3) seems facilitating tumor development through apoptosis inhibition. However, it suppresses metastasis in animal models, exhibiting different roles in tumor progression37. β-D mannuronic acid (BDM) is a MMP inhibitor, inhibiting MMP-2 and MMP-9 involved in invasion, metastasis, and angiogenesis38. BDM possesses anti-metastatic activity and inhibits tumor growth by suppressing inflammatory chemokine and tumor–promoting cytokines39. MMP-14 located on the cell surface, is a potential target to stop metastasis and a novel antibody-mediated MMP-14 blockade seems to limit hypoxia and metastasis in triple negative breast cancer (TNBC) models40. The progression from ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) exhibited up regulation of MMPs such as MMP-2, 11, 13, and 14 associated with invasion and ECM remodeling41. The MMPs along with cross-linking enzymes LOX (Lysil oxidase) facilitate collagen maturation, regulate expression and function of soluble factor like TGF-β which ultimately reciprocates through regulation of expression of many ECM proteins and modifying enzymes including LOXs42. Lox is a copper-dependent amine oxidase which initiates the intra- and intermolecular collagen crosslinking through oxidative deamination of specific lysine and hydroxylysine residues located in the telopeptide domains43. This crosslinking stiffens the matrix and promotes focal adhesions (Focal adhesion kinase level is increased), integrin clustering, PI3K signaling which ultimately facilitates ErbB2 – dependent breast tumor invasion11. The increased stiffness of matrix (measured by elastography) showed low response to neoadjuvant chemotherapy as compared to patients with soft breast carcinomas44,45. It is observed women with high mammographic density (MD) are more likely to develop BC as compared to women with low MD46. It seems down-regulation of LOXL4 promotes BC growth and lung metastasis in mice47. The LOXL2 protein catalyzes cross-linking of ECM components collagen and elastin and is involved in cancer progression and metastasis. The intracellular LOXL2 shows EMT induction and Snail-1 stabilization, and LOXL-2/Snail-1-mediated E-cadherin down-regulation promotes lung metastasis of BC without affecting ECM stiffness48. Collagen is the major scaffolding protein in stroma providing tensile strength to the tissue and its metabolism is dysregulated in cancer with increased expression and deposition49. The type I collagen is thought to provide barrier against tumor invasion; however enhanced collagen expression is observed with more incidence of metastasis50.
The enzyme collagen prolyl hydroxylase, required for collagen synthesis, is over expressed in BC tissues with poor prognosis3. Besides, the enzyme procollagen lysyl hydroxylase-2 involved in collagen synthesis, increases breast tumor stiffness, promotes metastatic tumors in lymph nodes and lungs. Matrix stiffness promotes tumor progression and invasion of ER+ type BC51. The hardened ECM drives invasion and metastasis through ERK1/2 signal up-regulation and JAK2/STAT5 signal down-regulation. The enzyme heparanase cleaves heparan sulfate, promoting tumor invasion and metastasis. ER stress during chemotherapy enhances the heparanase activity52. The MMTV-heparanase mice promoted growth and metastasis of breast tumor cells to lungs suggesting a role for heparanase in BC progression53. Elemene (extract of C urcuma erhizoma plant), is an anticarcinogenic phytochemical showing effects by down-regulating heparanase expression (potential target for heparanase)54. The heparin and nanoheparin derivatives show their anti-cancer activities by reducing BC cell proliferation and metastasis55. Loss of ECM integrity by plasmin facilitates cancer cell spread and plasmin-induced ECM degradation may be controlled by lipoprotein-A (competitive inhibitor of plasminogen)56–58. Vitamin C seems to be very important curbing tumor growth, and metastasis as ECM integrity requires vitamin C58.
The ECM proteins such as COL I, III, IV, VI, fibronectin, laminin 332, periostin, and vitronectin promote tumor progression and metastasis, whereas the proteins such as DMBT1, and SPARC suppress BC development and metastasis as reviewed by Zhu et al. (2014)3.
Different types of stromal cells that inhabit in the tumor microenvironment (TME), are immune cells, fibroblasts, adipocytes, endothelial cells and bonemarrow derived stem cells59. Tumor cells recruit tumor-associated macrophages (TAMs), which become proangiogenic by secreting VEGF-A which nourishes tumor cells and build a vessel network for their invasion. Hypoxia also induces macrophages to produce more VEGF and suppress immune response, promoting invasion60. Cancer-associated fibroblasts (CAFs) are involved in tumor development, progression, inflammation, metastasis, and build resistance to cancer therapy through secretion of hormones, cytokines, growth factors, etc. and cross-talk with other stromal cells, cancer cells, and ECM. CAFs facilitate the invasion through paracrine signaling with cancer cells. The cross-talk between CAFs and cancer cells enhances IGF1 secretion by CAFs and PAI-1 (Serpine1) activity in cancer cells61. These two molecules activate RhoA/ROCK signaling in cancer cells which increases cell scattering and invasion. Another study showed CAFs initially assembling an unfolded fibronectin matrix, later remodeled into a dense predominating collagen I matrix driven by MMPs62. This remodeling resulted in structural and mechanical changes in the stroma, promoting proangiogenic signaling and breast tumor invasion. CAFs can be potential therapeutic targets in BC63. Cancer cell proliferation and migration is induced by activated fibroblasts derived from endothelial-to-mesenchymal transformation64. The cancer associated adipocytes (CAAs) have a significant role in cancer progression, ECM remodeling, phenotype changes of CAFs, and resistance to cancer therapy65. They show tumor-modified phenotype with ability to modify cancer cell phenotype favoring metastasis66. Comparative gene expression profiling of myoepithelial cells of cancerous (DCIS) and normal breast tissue showed up regulation of several proteases (cathepsin F, K, and L, MMP2, and PRSS19), protease inhibitors (thrombospondin2, SERPING1, cytostatin C and TIMP3), and collagens like COL1A1, COL3A1, COL6A1 in DCIS tissue67,68.
Integrins, the primary receptors of MECs for ECM, act as sensors of epithelial microenvironment. They are the transmembrane glycoproteins present as heterodimers of α- and β- subunits. Total 8 β – subunits dimerize with 18 α – subunits to form around 24 distinct integrins which specifically bind to different ECM proteins69. Their altered expression seems to disorganize ECM and promotes metastasis70. Increased MEC proliferation occurs due to enhanced activity of integrin signaling (β1-, β5-, and β6- integrins) by co-activating the oncogenes which augment growth factor signaling. The β1 and β3 integrins play crucial role in BC progression and metastasis, hence therapy needs targeting these two integrins at once or their downstream cytokines like FAKs (focal adhesion kinases) and SFKs (Src family kinases) for effective treatment. One of the studies revealed integrin mediated BC invasion through integrin – uPAR (urokinase/plasminogen activator urokinase receptors) signaling which leads to FRA-1 (Fos-related antigen 1) phosphorylation and invasion71. The ECM protein vitronectin engagement via integrin and uPAR receptors, ends in activation of SRC and MAPK signaling which ultimately enhances FRA-1 phosphorylation. The FRA-1 (a member of AP-1 family of TFs) targets (which promote tumor cell proliferation, invasion and metastasis) include plasminogen activator, MMP-1, MMP-9, Clca2 (Chloride channel accessory2), adenosine receptor A2B, and miR221/222. Protein ECM1 is involved in angiogenesis, promoting TNBC migration and invasion72. Protein Hic-5 (focal adhesion scaffold/adaptor protein) promotes mammary duct formation. Focal adhesions of cells are attached to ECM and transduce signals from ECM to cell. Hic-5 is up-regulated in CAFs of BC, involved in EMT and invadopodia (F-actin rich protrusions of cancer cells) formation facilitating invasion, migration and metastasis73. The sustained directionality of tumor cells to a vessel is promoted by a chemotactic gradient of hepatocyte growth factor (HGF) produced from vessel endothelium. This directional streaming is possible by HGF/c-Met signaling pathway between endothelial cells and tumor cells; and c-Met inhibitors could be a potential target to block tumor cell streaming and metastasis74.
Clinical trials are going on intensively at present to target the stromal cells of TME for BC therapy in combination with cancerous cell targets, reviewed recently by Bahrami et al. (2018)59. To name a few drugs: drugs that target CAFs are chloroquine, metformin (targets lipid metabolism), anti-Met (targets glucose metabolism), celecoxib (COX-2 inhibitor), PD0332991 (cell cycle arrest), XAV 939 (β-catenin pathway inhibitor), SB431542 (TGF-β1receptor kinase inhibitor) etc. The drugs that target the immune cells are denosumab (Treg cell inhibitor), bisphosphonates (TAM inhibitor), indoximod (IDO pathway inhibitor) etc.
An anthracycline such as doxorubicin treatment of BC shows resistance to the drug mediated by ECM proteins as observed in the in vitro model75. Hence, probably combinatorial treatment with integrin signaling inhibitors would be more effective in BC therapy. ABL kinase inhibitors like imatinib, nilotinib, and GNF-5 impede the invadopodia formation, decrease ECM degradation, and impair the matrix proteolysis-dependent invasion as observed in the mouse xenograft model76.
The cytotoxic T-cells present in the TME kill the tumor cells. However, their infiltration is retarded by various factors. The chemokines such as CXCL-9, -10, -11(production is induced by IFN-γ and chemokine gradient is established) recruit the T-cells in the TME. The tumor cells produce the extracellular galectin-3 (a lectin that binds to glycans of glycoproteins in ECM), which binds to the glycoprotein IFN-γ and prevents it from inducing secretion of above chemokines, thus impedes the cytotoxic T-cell recruitment in the TME77. Immunotherapy targeting the galectin-3 would be better strategy to control tumor growth and invasion.
The TAMs (M2 phenotype) are protumoral in nature suppressing the adaptive immunity (suppresses CD8+ T cells), promoting angiogenesis, and matrix remodeling. The TAM can be a potential therapeutic target for BC therapy. Besides, the polarity switch from M2 to M1 phenotype (antitumoral function) could be a better strategy for treatment of cancer78,79.
MMP-14 was found to be a valid target to control tumor progression and metastasis in triple negative breast cancer. MMP-14 blockade by IgG3369 revealed decreased tumor neoangiogenesis and hypoxia80. MMP-11 can be a crucial tumor biomarker and a potential target for immunotherapy81.
High amount of hyaluronic acid (HA) present in TME seems to act as a physical barrier restricting the antibody and immune cell access to tumor cells82. It was observed pericellular matrix of HAhigh tumor cells restricted NK cell access and antibody- dependent cell mediated cytotoxicity (ADCC). The hyaluronidase (PEGPH20) treatment showed enhanced trastuzumab-dependent ADCC and NK cell mediated tumor growth inhibition in the in vivo system, proving an effective adjunctive therapy for HAhigh tumors82.
Benias et al. (2018)83 observed presence of fluid-filled interstitial space in the submucosa of many organs (which are subjected to intermittent compression) supported by thick collagen bundles. Similar structures may also present in breast tissues facilitating the metastasis of cancer cells as opposed to dense connective tissues acting as a barrier to migrating cancer cells.
The ECM constitutes a complex of structural proteins and its reorganization is essential during cancer progression. ECM proteins provide biochemical signals to induce EMT, promote metastasis progression of cancer to advanced stage. ECM remodeling enzymes like MMPs play an essential role in these processes. The TME, platelet-derived mitogens and chemokines, granulocytes and stromal cells help cancer cells achieve intravascular transit and metastasis to target site. In addition, various ECM proteins such as integrins, collagen and fibronectin engage in cell adhesion, invasion and metastasis. All these elements of the ECM are critical for cancer progression and hence targeting ECM is a prospective approach for targeted drug discovery and cancer therapy.
No data is associated with this article.
Views | Downloads | |
---|---|---|
F1000Research | - | - |
PubMed Central Data from PMC are received and updated monthly. | - | - |
Competing Interests: No competing interests were disclosed.
Reviewer Expertise: Cancer metastasis, tumor microenvironment, matrix metalloproteinases, immunotherapy
Competing Interests: No competing interests were disclosed.
Reviewer Expertise: Molecular breast oncology
Is the topic of the review discussed comprehensively in the context of the current literature?
Partly
Are all factual statements correct and adequately supported by citations?
Partly
Is the review written in accessible language?
Yes
Are the conclusions drawn appropriate in the context of the current research literature?
Partly
Competing Interests: No competing interests were disclosed.
Is the topic of the review discussed comprehensively in the context of the current literature?
Partly
Are all factual statements correct and adequately supported by citations?
Yes
Is the review written in accessible language?
Yes
Are the conclusions drawn appropriate in the context of the current research literature?
Yes
Competing Interests: No competing interests were disclosed.
Reviewer Expertise: Molecular biology, endocrinology of breast cancer
Is the topic of the review discussed comprehensively in the context of the current literature?
No
Are all factual statements correct and adequately supported by citations?
Partly
Is the review written in accessible language?
Yes
Are the conclusions drawn appropriate in the context of the current research literature?
Partly
References
1. Benias PC, Wells RG, Sackey-Aboagye B, Klavan H, et al.: Structure and Distribution of an Unrecognized Interstitium in Human Tissues.Sci Rep. 2018; 8 (1): 4947 PubMed Abstract | Publisher Full TextCompeting Interests: No competing interests were disclosed.
Reviewer Expertise: Cancer metastasis, tumor microenvironment, matrix metalloproteinases, immunotherapy
Alongside their report, reviewers assign a status to the article:
Invited Reviewers | |||
---|---|---|---|
1 | 2 | 3 | |
Version 2 (revision) 12 Jun 18 | read | read | |
Version 1 05 Mar 18 | read | read | read |
Provide sufficient details of any financial or non-financial competing interests to enable users to assess whether your comments might lead a reasonable person to question your impartiality. Consider the following examples, but note that this is not an exhaustive list:
Sign up for content alerts and receive a weekly or monthly email with all newly published articles
Already registered? Sign in
The email address should be the one you originally registered with F1000.
You registered with F1000 via Google, so we cannot reset your password.
To sign in, please click here.
If you still need help with your Google account password, please click here.
You registered with F1000 via Facebook, so we cannot reset your password.
To sign in, please click here.
If you still need help with your Facebook account password, please click here.
If your email address is registered with us, we will email you instructions to reset your password.
If you think you should have received this email but it has not arrived, please check your spam filters and/or contact for further assistance.
Comments on this article Comments (0)