Download PDF
Review  |  Open Access  |  18 Feb 2019

Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms

Views: 4506 |  Downloads: 1398 |  Cited:   12
J Cancer Metastasis Treat 2019;5:9.
10.20517/2394-4722.2019.01 |  © The Author(s) 2019.
Author Information
Article Notes
Cite This Article

Abstract

Postpartum involution is the process by which the lactating mammary gland returns to the pre-pregnant state after weaning. Expression of tumor-promotional collagen, upregulation of matrix metalloproteinases, infiltration of M2 macrophages, and remodeling of blood and lymphatic vasculature are all characteristics shared by the involuting mammary gland and breast tumor microenvironment. The tumor promotional nature of the involuting mammary gland is perhaps best evidenced by cases of postpartum breast cancer (PPBC), or those cases diagnosed within 10 years of most recent childbirth. Women with PPBC experience more aggressive disease and higher risk of metastasis than nulliparous patients and those diagnosed outside the postpartum window. Semaphorin 7a (SEMA7A), cyclooxygenase-2 (COX-2), and collagen are all expressed in the involuting mammary gland and, together, predict for decreased metastasis free survival in breast cancer. Studies investigating the role of these proteins in involution have been important for understanding their contributions to PPBC. Postpartum involution thus represents a valuable model for the identification of novel molecular drivers of PPBC and classical cancer hallmarks. In this review, we will highlight the similarities between involution and cancer in the mammary gland, and further define the contribution of SEMA7A/COX-2/collagen interplay to postpartum involution and breast tumor progression and metastasis.

Keywords

Postpartum involution, postpartum breast cancer, Semaphorin 7a, cyclooxygenase-2, collagen, metastasis

Breast cancer metastasis and postpartum breast cancer

In the United States, breast cancer remains the second leading cause of cancer related death in women, with the majority of these deaths resulting from metastatic disease. Breast cancer is a heterogeneous disease with five identified molecular sub-types: luminal-A, luminal-B, HER2-enriched, basal-like, and normal-like[1]. Treatments for hormone receptor positive and HER2 amplified cases include targeted therapies that, while initially successful, often result in the development of resistance, thereby increasing the likelihood of metastasis[2]. Breast cancers lacking estrogen receptor, progesterone receptor, and HER2 amplification are defined as triple negative breast cancers (TNBC) and account for 15%-20% of breast cancer diagnoses. Patients with TNBC not only have lower survival rates due to increased early metastasis, but also lack a targeted therapeutic option[3,4]. This evidence underscores the necessity of novel models for the identification of molecular drivers of breast cancer progression, therapy resistance, and metastasis.

Multiple epidemiological studies highlight the effect of pregnancy on breast cancer risk. While first pregnancy at an early age confers a lifelong protective effect against breast cancer, all women who have given birth undergo a transient period of increased risk[5-9]. Previously, women diagnosed within five years of most recent childbirth were found to be at higher risk for developing metastatic breast cancer than nulliparous women[5-12], a risk that persists even after adjustments have been made for variability in hormonal receptor status, HER2 status, age, histological grade, tumor size, node status, and year of diagnosis[10,13]. More recent results have revealed that the probability of metastasis is increased for women diagnosed with breast cancer between 0-5 and 5-10 years postpartum. Thus, we now define breast cancers diagnosed within 10 years of most recent childbirth as postpartum breast cancer (PPBC)[10,13]. By this definition, PPBC accounts for over half of all breast cancers diagnosed in women under age 40 in two independent cohorts[10,13]. Since patients with PPBC have significantly worse outcomes, regardless of numerous clinical parameters, it has been hypothesized that pro-tumorigenic changes in the breast tissue following pregnancy may persist for an extended period and accelerate PPBC progression. Consistent with this hypothesis, Asztalos et al.[14] identified a breast cancer associated genetic signature in the normal breast tissue of parous women that persists for up to 10 years after childbirth. Further evidence of a postpartum tumorigenic signature is supported by multiple pre-clinical models, where implantation of tumor cells into rodent mammary glands after weaning facilitates tumor cell growth, invasion, and metastasis. Additional studies of the mammary gland after lactation have revealed that these phenotypes are driven, in part, by mammary and tumor specific increases in pro-inflammatory cyclooxygenase-2 (COX-2), fibrillar collagen, semaphorin 7a (SEMA7A), bone marrow derived stromal and macrophage populations, lymphangiogenesis, and circulating estrogens[15-22]. In this review, we will explore some of the mechanisms by which post-lactational changes in the mammary gland facilitate breast cancer progression and metastasis, with a focus on the roles of collagen, COX-2, and SEMA7A in cell death, extracellular matrix (ECM) and vascular remodeling, and macrophage infiltration.

Pro-tumorigenic roles of involution associated programs

For a detailed review of the events that occur during embryonic and adult mammary gland development, see Macias and Hinck[23]. Briefly, mammary gland development begins during embryogenesis where ectodermal placodes invade into the mammary mesenchyme to form a rudimentary ductal tree[24]. This primitive structure persists until puberty where upregulation of growth hormone and estrogen coordinate ductal morphogenesis to form the extensive epithelial ductal network that fills the mammary fat pad. Mammary epithelial structures are bi-layered, meaning they consist of both luminal cells and basally restricted myoepithelial cells that contact the basement membrane[25]. Full differentiation of the mammary gland is not achieved until pregnancy, where progesterone and prolactin coordinate differentiation of the alveolar structures that are responsible for milk storage and secretion; in addition, continued branching morphogenesis occurs to prepare the gland for lactation[23,26]. During lactation, oxytocin stimulates the contraction of myoepithelial cells in response to suckling, resulting in milk delivery via the nipple[27]. When lactation ends, the gland must cease milk production and return to the pre-pregnant architecture through postpartum involution.

Postpartum involution involves the upregulation of tumor-promotional factors in the mammary epithelium and surrounding stroma. As primary components of the mammary ECM, collagen proteins, and the fibroblasts that produce and remodel them, have been extensively studied for their role in breast tumor progression and metastasis[28,29]. The 28 types of collagen proteins share a triple α-helix as part of their structure, and they can be broadly classified as fibrillar or non-fibrillar based on their assembly[30]. For additional information on collagen and its assembly, see Mouw et al.[30]. In this review, we will focus on fibrillar collagen, as this is the most abundant form in the mammary gland[31]. Collagen I expression is both spatially and temporally regulated, and its tight control is essential for the proper mechanosignaling required for normal mammary gland development and function[31]. In fact, dysregulation of the primary collagen I receptor, α2β1 integrin, can alter mammary ductal branching and promote tumor formation[32,33]. The contribution of collagen to tumor progression is emphasized by the prognostic value of collagen I mRNA in clinical outcomes of breast cancer[34] and evidence that increased collagen density promotes local breast cancer invasion and distant metastasis[35]. One way that collagen deposition is regulated during involution is by the pro-inflammatory enzyme COX-2.

COX enzymes were first identified as the targets of inhibition by aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs)[36]. COX enzymes exist in two isoforms, both of which bind to cell membranes where they catalyze the metabolism of free arachidonic acid (AA) to prostaglandins (PGs) via phospholipase A2[37,38]. First, COX enzymes rapidly catalyze the formation of an unstable intermediate, prostaglandin G2 (PGG2), from AA. PGG2 is then rapidly converted to prostaglandin H2 (PGH2) via COX-mediated peroxidase activity. Finally, specialized prostaglandin synthases result in the conversion of PGH2 to specific, biologically active PGs[37-39]. Though initially believed to function identically, later investigations revealed important differences between COX-1 and COX-2[40]. COX-1 is constitutively expressed as a regulator of tissue homeostasis[41], whereas COX-2 is not normally expressed in adult tissues, with the exception of the central nervous system[42], kidneys[43], and male reproductive organs[40,44]. Unlike COX-1, COX-2 expression is regulated by mitogens, hormones, and cytokines, and it is also correlated with cancer progression[45,46]. However, it is the product of COX-2 activity - PGE2 - that serves as the active effector of pro-tumorigenic signaling. Once synthesized, PGE2 can bind to specific E-prostanoid receptors on the cell surface to activate pathways associated with survival and inflammation[47]. COX-2/PGE2 signaling has been described in multiple models of cancer, including oral, breast, prostate, and colorectal, with documented roles in tumor initiation, invasion, immune evasion, cell survival, metastasis, vascular remodeling, cancer stem cells, and drug resistance; for further review, see Hashemi Goradel et al.[46] and Stasinopoulos et al.[48]. In addition to the feedback between COX-2 and collagen deposition in involution and in breast cancer[18], we have also published a connection between COX-2 and tumor cell invasion through expression of the neuronal guidance protein, SEMA7A[49].

SEMA7A, or CD108w, was first recognized for its expression on lymphocytes[50]; however, a role for SEMA7A in cancer was later identified through its association with Plexin-C1, the receptor for the viral homolog of SEMA7A. Engagement of Plexin C1 by viral semaphorin inhibits dendritic cell adhesion and motility via alterations to the actin cytoskeleton[51]. Similarly, in melanoma, SEMA7A/Plexin-C1 engagement inhibits cell migration via inactivation of the cofilin pathway[52]. Cofilin activation, which normally generates free actin filaments required for cell migration[53] is considered a major component of the metastatic cascade[54]. SEMA7A/Plexin-C1 mediated cofilin inactivation led to the identification of Plexin-C1 as a novel tumor suppressor[53]. Consistent with this finding, Plexin-C1 expression is frequently lost in melanoma[53]. Conversely, during development, SEMA7A signals through its other known receptor, β1-integrin, to promote axon outgrowth via activation of the mitogen activated protein kinase (MAP-K) pathway[55]. Unlike Plexin-C1, SEMA7A-mediated activation of β1-integrin has been shown to promote cancer progression. Our lab and others have shown that SEMA7A-β1-integrin binding promotes cell migration, invasion, metastasis, and neo-vasculogenesis of the blood and lymphatic vessels[49,56-58]. SEMA7A has also been implicated in multiple models of fibrosis (reviewed below), supporting additional roles in inflammation and fibrillar collagen deposition. The expression of SEMA7A, COX-2, and collagen during postpartum involution and their known roles in facilitating tumor progression suggest these molecules and their interplay may be important drivers of PPBC.

Pro-tumorigenic mechanisms of cell death during involution

Programmed cell death is essential for preventing the aberrant cellular phenotypes that arise when cells acquire DNA damage and mutations. Resistance to cell death may, consequently, result in the propagation of potentially harmful mutations that allow cells to bypass checkpoints meant to prevent unregulated growth and division. Resistance to cell death is thus defined by Hanahan and Weinberg[59] as one of the original hallmarks of cancer. For a detailed review of cell death and cancer, see Ichim and Tait[60]. In tumor cells, resistance to cell death plays an important role beyond tumor initiation, affecting drug-resistance, recurrence, and metastasis. Despite continuous advances in targeted chemotherapy, treatment efficacy tends to decline over time as a result of the ability of tumor cells to suppress apoptotic pathways, upregulate DNA repair, and genetically adapt to escape death. Tumor cells with repopulating capacity may also acquire the ability to resist death in response to chemotherapy, thereby constituting a major mechanism of cancer recurrence. Mechanisms by which tumor cells resist chemotherapy are further summarized by Al-Dimassi et al.[61]. Active survival signals from the mammary ECM are required to override the default programming of normal adherent cells to die. When these cells become detached, they die in the absence of ECM signaling by a specialized form of programmed cell death called anoikis[62]. Anoikis-resistance is a critical requirement for the successful metastasis of circulating tumor cells, a feature that may be shared by some of the cells that survive involution[63]. Postpartum involution is characterized by two major waves of apoptosis, culminating in the death of over half of the mammary epithelium[64]. As a minority of mammary epithelial cells (MECs) survive this process, postpartum involution represents an excellent model for studying the mechanisms by which cells acquire resistance to cell death in apoptotic environments. In this section, we will review the major mechanisms regulating cell death during postpartum involution and potential contributors to cell death resistance during tumorigenesis.

Postpartum involution occurs in two primary phases. In the first, or reversible phase, accumulation of milk protein, a process known as milk stasis, results in the detachment and shedding of secretory alveolar epithelial cells to the lumen. Milk stasis induces the first wave of cell death, in part, through the local expression of leukemia inhibitory factor (LIF). LIF is a primary activator of the master apoptotic regulator of involution - signal transducer and activator of transcription 3 (STAT3)[65]. Other important activators of STAT3 at the onset of postpartum involution include transforming growth factor-β (TGFβ), Janus kinase-1 (JAK-1), and Snail2 (SLUG)[66-68]. Activation of STAT3 promotes postpartum mammary gland involution by shifting the balance of pro- and anti-apoptotic signals in favor of programmed cell death. STAT3 coordinates the first wave of epithelial apoptosis through activation of pro-apoptotic Bcl-2 family members, upregulation of PI3K inhibitory subunits, and downregulation of MAP-K survival signaling[66]. STAT3 further promotes cell death during involution by mediating the formation of triglyceride-containing vacuoles. Triglycerides within these lysosomal-like vacuoles become metabolized into free fatty acids which interact with and distort lysosomal membranes. This results in the leakage of cathepsin proteases into the cytosol, and ultimately, lysosomal-mediated programmed cell death[69]. The role of STAT3 in regulating apoptosis during involution has recently been comprehensively reviewed by Hughes and Watson[66]. STAT3 has a well-defined role in cancer progression, with over 40% of breast cancers presenting with constitutive STAT3 activation. Because STAT3 activates multiple signaling pathways, aberrant activation can promote multiple changes associated with cancer, including altered cell cycle dynamics, EMT, angiogenesis, and interestingly, resistance to cell death[70,71]. Therapies targeting STAT3 may, thus, be of important therapeutic value for PPBC patients. For more about the role of STAT3 in breast cancer, see Segatto et al.[70].

Removal of the overwhelming number of dead cells from the gland requires the Rac-1 mediated switch of MECs from a secretory to a phagocytic phenotype - a process which is essential for proper remodeling in the second phase of involution[72]. Despite the massive wave of cell death that occurs during the first phase, if suckling resumes within this window (48-72 h, in mice), involution can be reversed, and lactation can proceed. This reversibility is due, in large part, to the expression of tissue inhibitors of metalloproteinases (TIMPs)[73]. TIMPs preserve the reversibility of gland regression by delaying irreversible tissue remodeling events through the inhibition of matrix metalloproteinases (MMPs)[74]. Interestingly, TIMP expression may also serve as an additional mechanism to regulate epithelial apoptosis, as TIMP3 has been implicated in the regulation of cell death in a tumor necrosis factor (TNF)-dependent manner[75]. The number of compensatory mechanisms that exist to activate apoptosis underscore the importance of postpartum involution to future rounds of successful lactation. At approximately day three of involution in mice, downregulation of TIMPs results in activation of the MMPs that degrade the mammary ECM, causing MECs to lose contact with their underlying basement membrane[73]. In the absence of pro-survival signaling from the ECM, detached MECs die by anoikis, thus comprising the second wave of cell death[62,73]. Similar to the first phase, clearance of dead cells during the second phase of involution is largely mediated by phagocytic MECs, with additional limited support from professional phagocytes. Milk fat globule epidermal growth factor 8, which works by recognizing phosphatidylserine on the outer leaflet of the plasma membrane of dying cells, is also critical for apoptotic cell clearance during the second phase[76].

What allows some cells to die and others to live during involution remains largely unanswered. To date, more than 50 mammary specific knockouts have been generated that exhibit alterations in postpartum involution - either delayed or premature - that are consistent with the proposed role for each molecule in the process. For example, activation of pro-survival pathways, such as Akt1, and/or deletion of death inducing genes, such as Bax, results in delayed involution, while deletion of Akt1 and anti-apoptotic Bcl-x results in premature involution[77-80]. See Radisky et al.[80] for an extensive review of these models. As pro-survival signals progressively decline and pro-apoptotic signals increase during the first phase of involution, the mammary basement membrane and ECM become the primary mediators of cell survival[81]. Yet, during the second phase, the basement membrane and ECM are degraded by proteases. Previously, SEMA7A mRNA expression was shown to increase in whole mammary extracts during the early phase of involution, and its expression was attributed to its immunomodulatory role[82]. Recently, we published that SEMA7A is expressed on Epcam+ MECs during the remodeling phase of involution[57]; however, the downstream mechanisms activated by epithelial SEMA7A are not well understood. Interestingly, SEMA7A is a ligand for β1-integrin, which is the receptor for ECM molecules that normally facilitate epithelial cell attachment. It is therefore possible that SEMA7A may activate β1-integrin signaling and provide a pro-survival mechanism to overcome anoikis. Furthermore, β1-integrin mediates the activation of known survival pathways including MAP-K and AKT, which affects survival via stabilization of NF-kB and expression of COX-2. Thus, SEMA7A may promote anoikis-resistance during involution via activation of β1-integrin in a manner that is independent of ECM; however, additional investigation is required.

The role of COX-2 during involution has also largely been attributed to its role in the modulation of the immune milieu[83]. Yet, COX-2 also contributes to cell survival mechanisms and collagen remodeling via PGE2, which promotes cell growth and proliferation, modulates collagen expression levels, and upregulates proteinase expression. Studies investigating the effects of postpartum involution in TNBC progression have revealed a feed-forward mechanism by which fibrillar collagen deposition during involution requires COX-2 expression. COX-2 further promotes increased fibrillar collagen[18], which is directly associated with tumor progression and poor prognosis in breast cancer patients[31,84]. Cooperation between fibrillar collagen and COX-2 may therefore contribute to the pro-tumorigenic nature of the involuting mammary gland. Similar to postpartum involution, wound healing is a biological process where cell death is accompanied by collagen remodeling. In the “phoenix rising” model of cell death, discovered in a model of wound healing and further characterized in cancer, dying cells send signals to stem and progenitor cells to increase their proliferation, thereby coordinating cell growth with death[85,86]. In dying cells, activation of caspases 3 and 7 results in the release of calcium-independent phospholipase-2, which increases the production and release of AA from cell membranes. Ultimately, COX-2 and PGE2 synthase convert free AA to PGE2, which increases stem and progenitor cell proliferation to promote tissue regeneration[85]. Though the phoenix rising model, to our knowledge, remains uninvestigated during mammary involution, there are numerous similarities between the wound healing program and postpartum involution, including apoptosis, clearance of damaged and dead cells, and activation of similar inflammatory programs. This suggests that this mechanism of coordinated cell death and tissue regeneration may be conserved in mammary development. Additionally, these mechanisms seem to be particularly important for the growth and survival of stem and progenitor cells in response to apoptotic stimuli, thus representing a mechanism of tumor recurrence in response to chemotherapy. The preceding data may help to further explain the role of collagen, COX-2, and SEMA7A in facilitating tumor progression following lactation.

Extracellular matrix and remodeling during postpartum involution and cancer

In their updated review of the hallmarks of cancer, Hanahan and Weinberg described the tumor microenvironment (TME) as a critical mediator of cancer progression due to its ability to supply cancer cells with signals that promote inflammation, induce angiogenesis, and confer resistance to cell death[87]. The TME consists of all the cells, ECM molecules, vasculature, and proteins that surround a tumor. The cross-talk between these components and the tumor can affect tumor growth, survival, metabolism, metastasis, response to treatment, and recurrence. The contributions of the TME to cancer progression are comprehensively reviewed in a recent special issue of Nature Reviews Cancer[88]. In this section, we will briefly discuss some of the similarities between the tissue microenvironment of the involuting mammary gland and the TME.

ECM fragments, which can be used as diagnostic markers of disease, are known contributors to cancer for their ability to participate in cell signaling events and modulate gene expression[31,89]. An investigation of the tumor-promotional aspects of involution revealed that tumor cells co-cultured with ECM isolated from involuting rat mammary gland promoted tumor cell invasion, whereas tumor cells co-cultured with ECM isolated from nulliparous rat mammary glands did not[90], suggesting that involution-derived ECM promotes tumor cell invasion and metastasis. In further support, orthotopic injection of breast cancer cells mixed with ECM isolated from the mammary glands of rats undergoing postpartum involution also confirmed that involution derived ECM promotes metastasis[90]. Additional investigations have confirmed that the involution microenvironment mirrors the typical breast TME based on their shared availability of ECM fragments, dramatic increases in fibrillar collagen, and increased MMP activity[90-92]. Specifically, studies of stomelysin-1 (MMP3), the primary active MMP during the tissue remodeling phase of involution, have characterized it as a potent mediator of EMT and other early oncogenic events in the mammary gland[93]. Furthermore, elevation of fibrillar collagen results in increased collagen crosslinking and ECM stiffening - characteristics known to accompany breast tumor progression[89,94-96]. Increased collagen deposition and cross-linking further promote tumor cell invasion and metastasis by providing a structural network for tumor cell migration[97].

Transforming growth factor β (TGFβ), well-known for its paradoxical role in breast tumor progression, is a primary regulator of collagen deposition in the mammary gland[98]. At approximately 8 h post weaning, TGFβ becomes activated and exerts tumor suppressive effects via activation of apoptotic programming[73,99]. Conversely, when normal cells begin to take on tumorigenic phenotypes, TGFβ promotes cancer progression by enhancing tumor cell survival and contributing to the maintenance of cancer stem cell populations[100]. TGFβ is also essential for wound healing, where it stimulates ECM deposition via fibroblast activation[101]. Interestingly, TGFβ dependent fibroblast activation during involution may be COX-2 dependent, as NSAID treatment decreases fibroblast activation in vivo. Furthermore, NSAIDs inhibit fibroblast mediated fibrillar collagen deposition during involution and in a model of PPBC[15]. Additionally, TGFβ is known to positively regulate SEMA7A in pulmonary fibrosis, where SEMA7A is critical for ECM deposition via activation of the PI3K/AKT pathway[102]. Given its role in ECM deposition, SEMA7A expression during the second phase of involution suggests it may also play a role in mammary gland tissue remodeling[57]. SEMA7A can affect ECM remodeling through its ability to recruit fibroblasts and immune cells to fibrotic sites, and it has been further implicated in fibrosis models in the liver, kidney, and in glial scar formation[103-108]. Contradictory to this role, when endogenously expressed on fibroblasts, SEMA7A can maintain fibroblast homeostasis and reduce pro-fibrotic markers[109], indicating context dependent roles for SEMA7A-mediated signaling. In cancer, fibrillar collagen coordinates upregulation of COX-2 on tumor cells, further promoting tumor cell invasion and metastasis[7], and we have published that COX-2 drives SEMA7A expression[49]. Thus, fibrillar collagen, COX-2, and SEMA7A may be a part of a feed-forward loop that ultimately results in cancer cell invasion and metastasis. Additional studies, however, are needed to better understand the hierarchy and cross-talk between these molecules in the context of postpartum involution and breast cancer progression.

Altering fibrillar collagen deposition provides a route for tumor cell migration and invasion, but also changes the signals received by cells from the surrounding environment. Signals from the ECM are communicated to cells by integrins. Integrins are heterodimeric (αβ) transmembrane receptors with 18 known α and 8 β integrin subunits, resulting in 24 possible heterodimeric integrin receptors[110]. Proper expression and signaling of integrins is essential for cell survival and adhesion, and integrin dysregulation can promote cancer via activation of pathways that affect survival, EMT, and migration[28,111-115]. Specifically, improper integrin signaling and expression can result in the loss of normal epithelial cell polarity and attachment, in addition to the over-activation of focal adhesion kinase and subsequent downstream signaling pathways that promote cell survival, migration, invasion, and ultimately metastasis[116]. While some integrin pairs are highly specific in their substrate recognition, others can recognize a number of substrates from the ECM, as well as foreign molecules such as snake venom, viral particles, and pathogens[117,118]. Alternative ligand-binding partners and/or differential integrin expression can elicit different signaling pathways; thus, when either ligand or integrin profiles are altered, cellular signaling pathways can become aberrantly activated or inhibited. Abnormal SEMA7A expression during involution and/or cancer may promote tumorigenesis via activation of β1-integrin and downstream pathways. SEMA7A binds to β1-integrin via the RGD binding site located on the SEMA domain; however, this site is buried in the crystal structure when SEMA7A is bound to Plexin C1. The ability to differentially regulate tumor progression could, therefore, be explained by binding of SEMA7A to its different receptors[119,120]. Further, while SEMA7A binds to α1β1-integrin on inflammatory macrophages[121], the a-binding partner needed for SEMA7A-mediated breast cancer progression is unknown. As reviewed above, one consequence of SEMA7A-β1-integrin signaling is fibrillar collagen deposition. Collagen can also activate integrins and modulate their associated signaling pathways, primarily through α2β1 integrin, which is often upregulated on cancer cells of epithelial origin[122]. Collagen binding to α2β1 integrin on tumor cells promotes cellular invasion, which helps cells navigate through the collagen I rich mammary TME and distant metastatic sites, such as the bone[122]. Interestingly, α2β1 integrin has been shown to increase COX-2 expression in intestinal epithelial cells[123], leading to activation of downstream signaling events associated with tumor promotion. While these results further support a link between COX-2 and collagen, α2β1 integrin has also been recognized as a metastasis suppressor in breast cancer. The complex interplay between integrins, collagen, COX-2, and SEMA7A is, thus, likely to be context-dependent, and additional studies are necessary to understand the role of this signaling axis in mediating tumor cell invasion and metastasis in breast cancer.

Macrophages in postpartum involution and PPBC

Macrophages are the phagocytic immune cells that mediate the removal of foreign pathogens, dead cells, and debris. Classically-activated macrophages, also known as M1 macrophages, are activated in response to pathogen-associated cytokines, most often IFN-γ and lipopolysaccharide. M1 macrophages are largely considered to be “anti-tumor” based on their expression of the pro-inflammatory cytokines, interleukin-1 (IL-1), IL-12, TNF-α, and inducible nitric oxide synthase - all of which have been shown to oppose tumor progression[124,125]. In contrast, Th2 family cytokines induce the maturation of alternatively-activated, or M2 macrophages, and cause their release of anti-inflammatory mediators that support tumor cell survival[21,126]. M2 macrophages promote tumor cell growth, invasion, and metastasis, via their secretion of IL-10, TGFβ, and MMPs. Though the M1/M2 system is useful for broadly classifying macrophages, this taxonomy fails to capture the plasticity and diversity characteristic of this cell type. For the purpose of this review, however, we will use M1 and M2 to broadly classify anti- and pro-tumor macrophages, respectively. Multiple studies support the role of macrophages as critical mediators of metastasis[21,127-129]. In models of gastric and breast cancer, M2 macrophages are recruited by tumor cells, where they activate MAP-K signaling to promote the motility of disseminating tumor cells[127]. Further, macrophages appear to be critical for the migration of the majority of ductal carcinoma in situ cells, as only 10% are motile when macrophages are absent[130]. The critical contribution of macrophages to tumor cell metastasis is further evidenced by studies in the MMTV-PyMT mouse model of breast cancer, where knockout of colony stimulating factor-1 (CSF-1), a secreted glycoprotein that induces the differentiation of hematopoietic stem cells to macrophages, correlates with a near complete elimination of tumor cell metastasis[21,131]. Macrophages, therefore, represent a diverse population of cells that can promote or inhibit tumor progression based on the context of their environment.

Macrophages are the primary immune cells present during mammary gland postpartum involution, and because of their role in facilitating tumor metastasis, represent a potential contribution to the highly metastatic nature of PPBC. Though known primarily for their phagocytic capacity, macrophages only play a minimal role in the clearance of apoptotic cells during involution[21,132,133]. Despite their limited role in phagocytosis, M2 macrophages are essential for the epithelial apoptosis and tissue remodeling characteristic of postpartum involution[134]. At the peak of apoptotic cell clearance, macrophages exist at relatively low levels, as MECs represent the primary phagocytes. At day 6 of involution, however, the peak of mammary tissue remodeling, M2 macrophages exist at 6 times the level of those in the nulliparous mammary gland, while classically-activated M1 macrophages remain at consistent levels throughout pregnancy, lactation, and gland regression[21]. F4/80, a general marker of mature mouse macrophages, marks more than the sum of M1 and M2 macrophages during involution, suggesting there are additional macrophage populations present in the involuting mammary gland[20]. Our lab has recently identified a population of macrophages that also express the lymphatic endothelial marker, podoplanin (PDPN)[57]. In culture, SEMA7A drives the expression of PDPN on macrophages and promotes their migration and adherence to lymphatic vessels[57]. Because macrophages have proven to be a critical part of the metastatic cascade by facilitating intravasation into tumor associated blood vessels[135], SEMA7A-mediated macrophage lymphatic mimicry may also facilitate intravasation into lymphatic vessels, providing another explanation for the high rates of metastasis associated with PPBC. This is further supported by the prognostic value of a combined genetic signature of CD68, PDPN, and SEMA7A in predicting decreased distant metastasis free survival in a cohort of 600 human breast cancer cases[57]. SEMA7A further regulates macrophages by serving as a strong activation factor for monocytes, promoting both chemotaxis and secretion of inflammatory cytokines, in addition to upregulation of granulocyte-macrophage CSF (GM-CSF), supporting an additional role for SEMA7A in macrophage differentiation[136].

Another important macrophage regulator during postpartum involution and breast cancer is COX-2. Previous studies in breast cancer models have shown that COX-2 expression increases with cancer stage, and its expression levels can indicate breast cancer progression, recurrence, and metastasis[137]. Recently, COX-2 expressing tumor associated macrophages (TAMs) have been shown to promote the metastatic potential of breast cancer cells via secretion of IL-6 and subsequent activation of AKT signaling in cancer cells[138]. Further, expression of COX-2 in stromal TAMs results in upregulation of COX-2 in breast cancer cells, thereby shifting polarization of local macrophages toward the M2 phenotype. In addition to its association with tumor promotional CD163+ TAMs, COX-2 expression in the stroma is further associated with increased collagen alignment in invasive breast cancer[139]. TAMs are known to associate with dense regions of collagen in breast cancer in the same way M2 macrophages associate with fibrillar collagen during involution. In the MMTV-PyMT model, macrophages associated with fibrillar collagen have been shown by intravital imaging to migrate across collagen fibers, suggesting that one mechanism by which macrophages promote metastasis is by supporting the migration of tumor cells across collagen networks[21,140]. SEMA7A, COX-2, and collagen all represent important effectors of macrophage-mediated tumor cell growth, survival, and metastasis. As macrophages are considered essential for successful metastasis, targeting the molecules responsible for alternative macrophage activation, survival, and chemotaxis may be critical for the successful treatment of metastatic disease.

Endothelial vessel formation

Blood and lymphatic vessels form two similar, yet distinct, organ systems that assemble into extensive networks throughout the body to support development and survival. Blood vessels provide tissues with oxygen and nutrients, while the primary functions of lymphatic vessels are immune cell trafficking and removal of excess interstitial fluid from tissues. Blood and lymphatic vessels are lined with blood endothelial cells and lymphatic endothelial cells, respectively, and are both surrounded by a thin layer of smooth muscle. Some lymphatic vessels have unique “button-like” junctions that differ from the more continuous “zipper-like” junctions of the blood vasculature and established lymphatic vessels. These specialized junctions are covered with a flap that opens and closes to allow fluid and cells to pass without affecting vascular integrity. Vascular networks are highly dynamic, expanding and retracting as tissues change in response to normal developmental processes or pathologies. Indeed, a widely accepted hallmark of cancer is the ability of tumors to induce angiogenesis, or the development of new blood vessels from existing vasculature[59]. Tumors must acquire pro-angiogenic abilities in order to grow beyond 1-2 mm3[141]; otherwise, the tumor will die by necrosis or apoptosis[142,143]. In rat mammary tissues, we observe an overall net increase in lymphatic vessel density (LVD) during involution when compared to a lactation timepoint[16]. In contrast, blood vessel density (BVD) drops dramatically after lactation, suggesting an initial period of regression before increasing in a manner similar to LVD[144]. During involution, the highest vasculature densities peak at day 10, followed by a slight decrease in the fully regressed gland [Figure 1]. This is consistent with published studies from our group and others describing increased pro-angiogenic and pro-lymphatic signaling during postpartum involution, and in postpartum tumors compared to non-postpartum controls[14,16,22,145]. Interestingly, fibrillar collagen, COX-2, and SEMA7A all have established roles in endothelial vessel formation. Studies using artificial collagen matrices have shown that collagen increases angiogenic responses from endothelial cells by providing the support needed for sustained endothelial cell growth and the formation of endothelial networks[146]. Angiogenesis can also be regulated by mechanical stiffness within the small microvessel environment[147], and breast tumors are often stiffer than neighboring normal tissues by up to 6-fold[84,148]. For a more comprehensive review on collagen and angiogenesis, see Fang et al.[149]. These studies demonstrate how the ECM in the TME can modulate vessel formation and alter the tumor’s blood supply.

Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms

Figure 1. BVD and LVD during mammary gland development. BVD measured as number of blood vessels per millimeter and LVD measured as number of lyve-1 positive vessels per millimeter in nulliparous (N), pregnant (P), lactating (L), involuting (days 2-10) and regressed (R), mammary glands (data adapted from Lyons et al.[16] and Ramirez et al.[144]). BVD: Blood vessel density; LVD: lymphatic vessel density

If a tumor outgrows its blood supply and loses its access to oxygen, it can become hypoxic. Under normal oxygen content conditions, termed “normoxia,” the transcriptional regulator hypoxia-inducible factor-1 (HIF-1) is unable to affect its targets; for further review of this topic, see Masson and Ratcliffe[150]. HIF-1 is comprised of two subunits - ARNT/HIF-1β, which is constitutively expressed, and HIF-1α, which is an oxygen-sensitive subunit. During normoxia, HIF-1α is hydroxylated by prolyl hydroxylase domain containing proteins, ubiquitinated by von-Hippel-Lindau protein, and rapidly degraded. During hypoxia, however, HIF-1α is not hydroxylated and degraded, but instead, translocates to the nucleus, heterodimerizes with ARNT, and induces transcription of its target genes. HIF-1 affects a variety of targets, including pro-angiogenic genes like vascular endothelial growth factor-A (VEGF-A)[151]. In breast cancer cells, COX-2 can induce inflammation-associated HIF-1 activity, resulting in the expression of pro-angiogenic genes[48]. Further, HIF-1 and COX-2 maintain a positive feedback loop, as HIF-1 can also induce expression of COX-2[152]. Utilizing NSAIDs to target COX-2 activity can inhibit angiogenesis, demonstrating the potential for therapeutic intervention[152-154]. Corroborating our findings that SEMA7A may be involved in COX-2 associated pathways, HIF-1 can directly upregulate SEMA7A in endothelial cells[155]. SEMA7A can elicit the release of pro-inflammatory cytokines and cause increased endothelial barrier permeability[156,157]. SEMA7A can also promote angiogenesis in a hypoxia-independent manner in murine mammary carcinoma and in the cornea by stimulating macrophages to produce pro-angiogenic molecules, such as CXCL2/MIP-2[158] and VEGF-A[159].

In addition to their roles in angiogenesis of the blood vasculature, collagen, COX-2, and SEMA7A also have known roles in lymphangiogenesis. In breast cancer, increased LVD, lymph node involvement, and lymph vessel invasion are predictive of higher risk of metastasis; further, increased LVD and lymph node metastasis are commonly observed in PPBC[16]. While the role of collagen in lymphangiogenesis has not been extensively characterized, one study has shown that collagen I increases lymphangiogenesis and angiogenesis in mouse embryoid bodies under hypoxic conditions[160]. Therefore, it is plausible that other fibrillar collagens may contribute to lymphangiogenesis during mammary tumorigenesis. COX-2/PGE2 signaling also promotes production of pro-angiogenic VEGF-A and pro-lymphangiogenic VEGF-C and VEGF-D[161]; further, lymphangiogenesis during wound healing is dependent on COX-2 activity[162]. Moreover, COX-2 has been implicated in lymphangiogenesis in other cancer types, including cervical and gastric[163,164]. We published that inhibition of COX-2 with celecoxib and NSAIDs results in decreased LVD, tumor cell invasion into lymphatics, and metastasis during PPBC[16]. For more detailed information on COX-2 and lymphangiogenesis in breast cancer, see Lala et al.[165]. Finally, SEMA7A has a functional role in lymphatic vessel modulation, as we recently published that SEMA7A promotes tumor-associated lymphangiogenesis via macrophage-mediated lymphatic vessel remodeling during postpartum involution and breast cancer[57]. These findings suggest an additional mechanism by which SEMA7A, COX-2, and collagen promote tumor progression and metastasis.

Clinical relevance/conclusion

Women with postpartum breast cancer face a disease that carries three times the rate of metastasis and death relative to women who have never been pregnant and those diagnosed outside of the postpartum window. One physiological event these women have in common is postpartum involution - a process that results in the upregulation and activation of tumor-promotional factors in MECs and the mammary stroma. Identification of the genetic engines that drive PPBC is critical to the development of targeted therapies for postpartum patients. In this review, we have highlighted potential roles for collagen, COX-2, and SEMA7A in driving some of the pro-metastatic aspects of involution [Figure 2]. Previously published results indicate overall survival is generally decreased for breast cancer patients with high collagen, COX-2, and SEMA7A expression, suggesting that these mechanisms are important mediators of breast cancer metastasis[18,49]. Interestingly, while individual expression of each molecule does not predict for metastasis using KM Plotter analysis[166], the combination of high SEMA7A, COX-2, and COL1A1 mRNA expression results in significantly decreased distant metastasis free survival for breast cancer patients in this dataset [Figure 3]. Thus, studies rooted in understanding the contributions of postpartum involution associated programs to breast cancer metastasis are likely to also be applicable to general breast cancer metastasis, and perhaps to other cancer types.

Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms

Figure 2. Graphical summary of pro-tumorigenic and metastatic roles of COX-2 and SEMA7A during postpartum involution and postpartum breast cancer. SEMA7A: Semaphorin 7a; COX-2: cyclooxygenase-2

Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms

Figure 3. Distant metastasis free survival is decreased with high levels of expression of SEMA7A, COX-2, and COL1A1 signature. DMFS analysis using KmPlot (n = 664) *P < 0.05

Based on the cooperation between SEMA7A, COX-2, and collagen, a multi-targeted therapy to affect the individual molecules and their interplay would likely be more effective than targeting one, alone. The potential of COX-2 as a therapeutic treatment has been investigated in multiple models of cancer. In fact, the COX-2 inhibitor, celecoxib, has been successful in the treatment of a specific type of colorectal cancer - familial adenomatous polyposis - in both adults and children[167,168]. Targeting COX-2 in breast cancer, by celecoxib or other NSAIDs, may inhibit tumor cell dissemination by reducing the expression of tumor-promotional collagen. Targeting SEMA7A in conjunction with already established therapies, such as NSAIDs, may also increase the efficacy of these treatments in women with breast cancer. Ideally, the characterization of tumor-promotional factors in the postpartum mammary gland may also lead to preventative therapies aimed at reducing the risk for PPBC. NSAIDs may further represent a safe candidate for preventative therapy during involution via inhibition of COX-2 mediated collagen upregulation and alternative macrophage activation. The topics covered herein highlight both the potential contribution of the SEMA7A/COX-2/Collagen relationship to PPBC, and the importance of PPBC models to the discovery of new molecules and pathways that can be exploited as novel therapeutics.

Declarations

Acknowledgments

The authors would like to give special thanks to Alan Elder and Chloe Young for their critical contributions to the editing process and PA Masso-Welch for providing the raw data from published studies[144] for Figure 1.

Authors’ contributions

Conception and design: Wallace TR, Tarullo SE, Lyons TR

Drew figures: Tarullo SE, Lyons TR

Manuscript writing/editing: Wallace TR, Tarullo SE, Crump LS, Lyons TR

Read and approved the final manuscript: Wallace TR, Tarullo SE, Crump LS, Lyons TR

Availability of data and materials

Not applicable.

Financial support and sponsorship

This work was supported by American Cancer Society (RSG-16-171-010CSM); National Cancer Institute (R01 CA211696-01A1); National Cancer Institute (R01 CA211696-02S1); and National Cancer Institute (F31 CA236140).

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2019.

REFERENCES

1. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, et al. Molecular portraits of human breast tumours. Nature 2000;406:747-52.

2. Masoud V, Pagès G. Targeted therapies in breast cancer: new challenges to fight against resistance. World J Clin Oncol 2017;8:120-34.

3. Liedtke C, Mazouni C, Hess KR, Andre F, Tordai A, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol 2008;26:1275-81.

4. Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer registry. Cancer 2007;109:1721-8.

5. Albrektsen G, Heuch I, Kvale G. The short-term and long-term effect of a pregnancy on breast cancer risk: a prospective study of 802,457 parous Norwegian women. Br J Cancer 1995;72:480-4.

6. Lambe M, Hsieh C, Trichopoulos D, Ekbom A, Pavia M, et al. Transient increase in the risk of breast cancer after giving birth. N Engl J Med 1994;331:5-9.

7. Lyons TR, Schedin PJ, Borges VF. Pregnancy and breast cancer: when they collide. J Mammary Gland Biol Neoplasia 2009;14:87-98.

8. Nichols HB, Schoemaker MJ, Cai J, Xu J, Wright LB, et al. Breast cancer risk after recent childbirth: a pooled analysis of 15 prospective studies. Ann Intern Med 2018; doi: 10.7326/M18-1323.

9. Schedin P. Pregnancy-associated breast cancer and metastasis. Nat Rev Cancer 2006;6:281-91.

10. Callihan EB, Gao D, Jindal S, Lyons TR, Manthey E, et al. Postpartum diagnosis demonstrates a high risk for metastasis and merits an expanded definition of pregnancy-associated breast cancer. Breast Cancer Res Treat 2013;138:549-59.

11. Leon DA, Carpenter LM, Broeders MJ, Gunnarskog J, Murphy MF. Breast cancer in Swedish women before age 50: evidence of a dual effect of completed pregnancy. Cancer Causes Control 1995;6:283-91.

12. Janerich DT, Hoff MB. Evidence for a crossover in breast cancer risk factors. Am J Epidemiol 1982;116:737-42.

13. Goddard ET, Bassale S, Schedin T, Jindal S, Johnston J, et al. Association between postpartum breast cancer diagnosis and metastasis and the clinical features underlying risk. JAMA Netw Open 2019;2:e186997.

14. Asztalos S, Gann PH, Hayes MK, Nonn L, Beam CA, et al. Gene expression patterns in the human breast after pregnancy. Cancer Prev Res (Phila) 2010;3:301-11.

15. Guo Q, Minnier J, Burchard J, Chiotti K, Spellman P, et al. Physiologically activated mammary fibroblasts promote postpartum mammary cancer. JCI Insight 2017;2:e89206.

16. Lyons TR, Borges VF, Betts CB, Guo Q, Kapoor P, et al. Cyclooxygenase-2-dependent lymphangiogenesis promotes nodal metastasis of postpartum breast cancer. J Clin Invest 2014;124:3901-12.

17. McCready J, Arendt LM, Rudnick JA, Kuperwasser C. The contribution of dynamic stromal remodeling during mammary development to breast carcinogenesis. Breast Cancer Res 2010;12:205.

18. Lyons TR, O’Brien J, Borges VF, Conklin MW, Keely PJ, et al. Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2. Nat Med 2011;17:1109-15.

19. Martinson HA, Jindal S, Durand-Rougely C, Borges VF, Schedin P. Wound healing-like immune program facilitates postpartum mammary gland involution and tumor progression. Int J Cancer 2015;136:1803-13.

20. O’Brien J, Lyons T, Monks J, Lucia MS, Wilson RS, et al. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. Am J Pathol 2010;176:1241-55.

21. O’Brien J, Schedin P. Macrophages in breast cancer: do involution macrophages account for the poor prognosis of pregnancy-associated breast cancer? J Mammary Gland Biol Neoplasia 2009;14:145-57.

22. Gupta PB, Proia D, Cingoz O, Weremowicz J, Naber SP, et al. Systemic stromal effects of estrogen promote the growth of estrogen receptor-negative cancers. Cancer Res 2007;67:2062-71.

23. Macias H, Hinck L. Mammary gland development. Wiley Interdiscip Rev Dev Biol 2012;1:533-57.

24. Balinsky BI. On the prenatal growth of the mammary gland rudiment in the mouse. J Anat 1950;84:227-35.

25. Monaghan P, Perusinghe NP, Cowen P, Gusterson BA. Peripubertal human breast development. Anat Rec 1990;226:501-8.

26. Oakes SR, Hilton HN, Ormandy CJ. The alveolar switch: coordinating the proliferative cues and cell fate decisions that drive the formation of lobuloalveoli from ductal epithelium. Breast Cancer Res 2006;8:207.

27. Haaksma CJ, Schwartz RJ, Tomasek JJ. Myoepithelial cell contraction and milk ejection are impaired in mammary glands of mice lacking smooth muscle alpha-actin. Biol Reprod 2011;85:13-21.

28. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 2012;196:395-406.

29. Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep 2014;15:1243-53.

30. Mouw JK, Ou G, Weaver VM. Extracellular matrix assembly: a multiscale deconstruction. Nat Rev Mol Cell Biol 2014;15:771-85.

31. Schedin P, Keely PJ. Mammary gland ECM remodeling, stiffness, and mechanosignaling in normal development and tumor progression. Cold Spring Harb Perspect Biol 2011;3:a003228.

32. Keely PJ, Fong AM, Zutter MM, Santoro SA. Alteration of collagen-dependent adhesion, motility, and morphogenesis by the expression of antisense alpha 2 integrin mRNA in mammary cells. J Cell Sci 1995;108:595-607.

33. Chen J, Diacovo TG, Grenache DG, Santoro SA, Zutter MM. The alpha(2) integrin subunit-deficient mouse: a multifaceted phenotype including defects of branching morphogenesis and hemostasis. Am J Pathol 2002;161:337-44.

34. van ‘t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002;415:530-6.

35. Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, et al. Collagen density promotes mammary tumor initiation and progression. BMC medicine 2008;6:11.

36. Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol 1971;231:232-5.

37. Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid biology. Science 2001;294:1871-5.

38. Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 2000;69:145-82.

39. Cha YI, Solnica-Krezel L, DuBois RN. Fishing for prostanoids: deciphering the developmental functions of cyclooxygenase-derived prostaglandins. Dev Biol 2006;289:263-72.

40. Xie WL, Chipman JG, Robertson DL, Erikson RL, Simmons DL. Expression of a mitogen-responsive gene encoding prostaglandin synthase is regulated by mRNA splicing. Proc Natl Acad Sci U S A 1991;88:2692-6.

41. Yokoyama C, Takai T, Tanabe T. Primary structure of sheep prostaglandin endoperoxide synthase deduced from cDNA sequence. FEBS Lett 1988;231:347-51.

42. Snipes JA, Kis B, Shelness GS, Hewett JA, Busija DW. Cloning and characterization of cyclooxygenase-1b (putative cyclooxygenase-3) in rat. J Pharmacol Exp Ther 2005;313:668-76.

43. Adegboyega PA, Ololade O. Immunohistochemical expression of cyclooxygenase-2 in normal kidneys. Appl Immunohistochem Mol Morphol 2004;12:71-4.

44. Kirschenbaum A, Liotta DR, Yao S, Liu XH, Klausner AP, et al. Immunohistochemical localization of cyclooxygenase-1 and cyclooxygenase-2 in the human fetal and adult male reproductive tracts. J Clin Endocrinol Metab 2000;85:3436-41.

45. FitzGerald GA. COX-2 and beyond: approaches to prostaglandin inhibition in human disease. Nat Rev Drug Discov 2003;2:879-90.

46. Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: a review. J Cell Physiol 2018; doi: 10.1002/jcp.27411.

47. Yang H, Chen C. Cyclooxygenase-2 in synaptic signaling. Curr Pharm Des 2008;14:1443-51.

48. Stasinopoulos I, O’Brien DR, Bhujwalla ZM. Inflammation, but not hypoxia, mediated HIF-1alpha activation depends on COX-2. Cancer Biol Ther 2009;8:31-5.

49. Black SA, Nelson AC, Gurule NJ, Futscher BW, Lyons TR. Semaphorin 7a exerts pleiotropic effects to promote breast tumor progression. Oncogene 2016;35:5170-8.

50. Cerny J, Stockinger H, Horejsi V. Noncovalent associations of T lymphocyte surface proteins. Eur J Immunol 1996;26:2335-43.

51. Walzer T, Galibert L, Comeau MR, De Smedt T. Plexin C1 engagement on mouse dendritic cells by viral semaphorin A39R induces actin cytoskeleton rearrangement and inhibits integrin-mediated adhesion and chemokine-induced migration. J Immunol 2005;174:51-9.

52. Scott GA, McClelland LA, Fricke AF, Fender A. Plexin C1, a receptor for semaphorin 7a, inactivates cofilin and is a potential tumor suppressor for melanoma progression. J Invest Dermatol 2009;129:954-63.

53. Wang W, Eddy R, Condeelis J. The cofilin pathway in breast cancer invasion and metastasis. Nat Rev Cancer 2007;7:429-40.

54. Ono S. Mechanism of depolymerization and severing of actin filaments and its significance in cytoskeletal dynamics. Int Rev Cytol 2007;258:1-82.

55. Pasterkamp RJ, Peschon JJ, Spriggs MK, Kolodkin AL. Semaphorin 7A promotes axon outgrowth through integrins and MAPKs. Nature 2003;424:398-405.

56. Scott GA, McClelland LA, Fricke AF. Semaphorin 7a promotes spreading and dendricity in human melanocytes through beta1-integrins. J Invest Dermatol 2008;128:151-61.

57. Elder AM, Tamburini BAJ, Crump LS, Black SA, Wessells VM, et al. Semaphorin 7A promotes macrophage-mediated lymphatic remodeling during postpartum mammary gland involution and in breast cancer. Cancer Res 2018;78:6473-85.

58. Ma B, Herzog EL, Lee CG, Peng X, Lee CM, et al. Role of chitinase 3-like-1 and semaphorin 7a in pulmonary melanoma metastasis. Cancer Res 2015;75:487-96.

59. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70.

60. Ichim G, Tait SW. A fate worse than death: apoptosis as an oncogenic process. Nat Rev Cancer 2016;16:539-48.

61. Al-Dimassi S, Abou-Antoun T, El-Sibai M. Cancer cell resistance mechanisms: a mini review. Clin Transl Oncol 2014;16:511-6.

62. Vachon PH. Integrin signaling, cell survival, and anoikis: distinctions, differences, and differentiation. J Signal Transduct 2011;2011:738137.

63. Kim YN, Koo KH, Sung JY, Yun UJ, Kim H. Anoikis resistance: an essential prerequisite for tumor metastasis. Int J Cell Biol 2012;2012:306879.

64. Lund LR, Romer J, Thomasset N, Solberg H, Pyke C, et al. Two distinct phases of apoptosis in mammary gland involution: proteinase-independent and -dependent pathways. Development 1996;122:181-93.

65. Schere-Levy C, Buggiano V, Quaglino A, Gattelli A, Cirio MC, et al. Leukemia inhibitory factor induces apoptosis of the mammary epithelial cells and participates in mouse mammary gland involution. Exp Cell Res 2003;282:35-47.

66. Hughes K, Watson CJ. The multifaceted role of STAT3 in mammary gland involution and breast cancer. Int J Mol Sci 2018;19:E1695.

67. Castillo-Lluva S, Hontecillas-Prieto L, Blanco-Gomez A, Del Mar Saez-Freire M, Garcia-Cenador B, et al. A new role of SNAI2 in postlactational involution of the mammary gland links it to luminal breast cancer development. Oncogene 2015;34:4777-90.

68. Sakamoto K, Wehde BL, Yoo KH, Kim T, Rajbhandari N, et al. Janus kinase 1 is essential for inflammatory cytokine signaling and mammary gland remodeling. Mol Cell Biol 2016;36:1673-90.

69. Sargeant TJ, Lloyd-Lewis B, Resemann HK, Ramos-Montoya A, Skepper J, et al. Stat3 controls cell death during mammary gland involution by regulating uptake of milk fat globules and lysosomal membrane permeabilization. Nature cell biology 2014;16:1057-68.

70. Segatto I, Baldassarre G, Belletti B. STAT3 in breast cancer onset and progression: a matter of time and context. Int J Mol Sci 2018;19:E2818.

71. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 2009;9:798-809.

72. Akhtar N, Li W, Mironov A, Streuli Charles H. Rac1 controls both the secretory function of the mammary gland and its remodeling for successive gestations. Developmental Cell 2016;38:522-35.

73. Watson CJ. Involution: apoptosis and tissue remodelling that convert the mammary gland from milk factory to a quiescent organ. Breast Cancer Res 2006;8:203.

74. Green KA, Lund LR. ECM degrading proteases and tissue remodelling in the mammary gland. Bioessays 2005;27:894-903.

75. Hojilla CV, Jackson HW, Khokha R. TIMP3 regulates mammary epithelial apoptosis with immune cell recruitment through differential TNF dependence. PLoS One 2011;6:e26718.

76. Hanayama R, Nagata S. Impaired involution of mammary glands in the absence of milk fat globule EGF factor 8. Proc Natl Acad Sci U S A 2005;102:16886-91.

77. Schorr K, Li M, Bar-Peled U, Lewis A, Heredia A, et al. Gain of Bcl-2 is more potent than bax loss in regulating mammary epithelial cell survival in vivo. Cancer Res 1999;59:2541-5.

78. Walton KD, Wagner KU, Rucker EB 3rd, Shillingford JM, Miyoshi K, et al. Conditional deletion of the bcl-x gene from mouse mammary epithelium results in accelerated apoptosis during involution but does not compromise cell function during lactation. Mech Dev 2001;109:281-93.

79. Schwertfeger KL, Richert MM, Anderson SM. Mammary gland involution is delayed by activated Akt in transgenic mice. Mol Endocrinol 2001;15:867-81.

80. Radisky DC, Hartmann LC. Mammary involution and breast cancer risk: transgenic models and clinical studies. J Mammary Gland Biol Neoplasia 2009;14:181-91.

81. Li M, Liu X, Robinson G, Bar-Peled U, Wagner KU, et al. Mammary-derived signals activate programmed cell death during the first stage of mammary gland involution. Proc Natl Acad Sci U S A 1997;94:3425-30.

82. Clarkson RW, Wayland MT, Lee J, Freeman T, Watson CJ. Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res 2004;6:R92-109.

83. Fornetti J, Martinson HA, Betts CB, Lyons TR, Jindal S, et al. Mammary gland involution as an immunotherapeutic target for postpartum breast cancer. J Mammary Gland Biol Neoplasia 2014;19:213-28.

84. Acerbi I, Cassereau L, Dean I, Shi Q, Au A, et al. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol (Camb) 2015;7:1120-34.

85. Li F, Huang Q, Chen J, Peng Y, Roop DR, et al. Apoptotic cells activate the “phoenix rising” pathway to promote wound healing and tissue regeneration. Sci Signal 2010;3:ra13.

86. Huang Q, Li F, Liu X, Li W, Shi W, et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med 2011;17:860-6.

87. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74.

88. Nature reviews Cancer. The tumor microenvironment. Available from: https://www.nature.com/collections/khylqkxqbr/content/reviews. [Last accessed on 1 Feb 2019].

89. Provenzano PP, Inman DR, Eliceiri KW, Keely PJ. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK-ERK linkage. Oncogene 2009;28:4326-43.

90. McDaniel SM, Rumer KK, Biroc SL, Metz RP, Singh M, et al. Remodeling of the mammary microenvironment after lactation promotes breast tumor cell metastasis. Am J Pathol 2006;168:608-20.

91. Bemis LT, Schedin P. Reproductive state of rat mammary gland stroma modulates human breast cancer cell migration and invasion. Cancer Res 2000;60:3414-8.

92. O’Brien J, Hansen K, Barkan D, Green J, Schedin P, et al. Non-steroidal anti-inflammatory drugs target the pro-tumorigenic extracellular matrix of the postpartum mammary gland. Int J Dev Biol 2011;55:745-55.

93. Clarke R. Stromelysin-1 promotes mammary carcinogenesis. Breast Cancer Res 1999;1:66605.

94. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 2005;8:241-54.

95. Walker RA. The complexities of breast cancer desmoplasia. Breast Cancer Res 2001;3:143-5.

96. Kreike B, van Kouwenhove M, Horlings H, Weigelt B, Peterse H, et al. Gene expression profiling and histopathological characterization of triple-negative/basal-like breast carcinomas. Breast Cancer Res 2007;9:R65.

97. Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, et al. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med 2006;4:38.

98. Qiuchen G, Courtney B, Nathan P, Elizabeth M, Pepper S. Mammary gland involution provides a unique model to study the TGF-beta cancer paradox. J Clin Med 2017;6:10.

99. Nguyen AV, Pollard JW. Transforming growth factor beta3 induces cell death during the first stage of mammary gland involution. Development 2000;127:3107-18.

100. Imamura T, Hikita A, Inoue Y. The roles of TGF-β signaling in carcinogenesis and breast cancer metastasis. Breast Cancer 2012;19:118-24.

101. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-beta: the master regulator of fibrosis. Nat Rev Nephrol 2016;12:325-38.

102. Kang HR, Lee CG, Homer RJ, Elias JA. Semaphorin 7A plays a critical role in TGF-beta1-induced pulmonary fibrosis. J Exp Med 2007;204:1083-93.

103. Gan Y, Reilkoff R, Peng X, Russell T, Chen Q, et al. Role of semaphorin 7a signaling in transforming growth factor β1-induced lung fibrosis and scleroderma-related interstitial lung disease. Arthritis Rheum 2011;63:2484-94.

104. Reilkoff RA, Peng H, Murray LA, Peng X, Russell T, et al. Semaphorin 7a+ regulatory T cells are associated with progressive idiopathic pulmonary fibrosis and are implicated in transforming growth factor-β1-induced pulmonary fibrosis. Am J Respir Crit Care Med 2013;187:180-8.

105. Esnault S, Kelly EA, Johansson MW, Liu LY, Han ST, et al. Semaphorin 7A is expressed on airway eosinophils and upregulated by IL-5 family cytokines. Clin Immunol 2014;150:90-100.

106. De Minicis S, Rychlicki C, Agostinelli L, Saccomanno S, Trozzi L, et al. Semaphorin 7A contributes to TGF-β-mediated liver fibrogenesis. Am J Pathol 2013;183:820-30.

107. Kopp MA, Brommer B, Gatzemeier N, Schwab JM, Pruss H. Spinal cord injury induces differential expression of the profibrotic semaphorin 7A in the developing and mature glial scar. Glia 2010;58:1748-56.

108. Lee Y, Blount KL, Dai F, Thompson S, Scher JK, et al. Semaphorin 7A in circulating regulatory T cells is increased in autosomal-dominant polycystic kidney disease and decreases with tolvaptan treatment. Clin Exp Nephrol 2018;22:906-16.

109. Esnault S, Torr EE, Bernau K, Johansson MW, Kelly EA, et al. Endogenous semaphorin-7A impedes human lung fibroblast differentiation. PLoS One 2017;12:e0170207.

110. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell 2002;110:673-87.

111. Barkan D, El Touny LH, Michalowski AM, Smith JA, Chu I, et al. Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res 2010;70:5706-16.

112. Eke I, Cordes N. Focal adhesion signaling and therapy resistance in cancer. Semin Cancer Biol 2015;31:65-75.

113. Jiang WG, Sanders AJ, Katoh M, Ungefroren H, Gieseler F, et al. Tissue invasion and metastasis: molecular, biological and clinical perspectives. Semin Cancer Biol 2015;35 Suppl:S244-S75.

114. Narunsky L, Oren R, Bochner F, Neeman M. Imaging aspects of the tumor stroma with therapeutic implications. Pharmacol Ther 2014;141:192-208.

115. Seguin L, Desgrosellier JS, Weis SM, Cheresh DA. Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol 2015;25:234-40.

116. Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol 2004;5:816-26.

117. Hamidi H, Ivaska J. Every step of the way: integrins in cancer progression and metastasis. Nat Rev Cancer 2018;18:533-48.

118. Humphries JD, Byron A, Humphries MJ. Integrin ligands at a glance. J Cell Sci 2006;119:3901-3.

119. Liu H, Juo ZS, Shim AH, Focia PJ, Chen X, et al. Structural basis of semaphorin-plexin recognition and viral mimicry from Sema7A and A39R complexes with PlexinC1. Cell 2010;142:749-61.

120. Jongbloets BC, Lemstra S, Schellino R, Broekhoven MH, Parkash J, et al. Stage-specific functions of semaphorin7A during adult hippocampal neurogenesis rely on distinct receptors. Nat Commun 2017;8:14666.

121. Suzuki K, Okuno T, Yamamoto M, Pasterkamp RJ, Takegahara N, et al. Semaphorin 7A initiates T-cell-mediated inflammatory responses through alpha1beta1 integrin. Nature 2007;446:680-4.

122. Naci D, Vuori K, Aoudjit F. Alpha2beta1 integrin in cancer development and chemoresistance. Semin Cancer Biol 2015;35:145-53.

123. Broom OJ, Massoumi R, Sjolander A. Alpha2beta1 integrin signalling enhances cyclooxygenase-2 expression in intestinal epithelial cells. J Cell Physiol 2006;209:950-8.

124. Esquivel-Velazquez M, Ostoa-Saloma P, Palacios-Arreola MI, Nava-Castro KE, Castro JI, et al. The role of cytokines in breast cancer development and progression. J Interferon Cytokine Res 2015;35:1-16.

125. Vannini F, Kashfi K, Nath N. The dual role of iNOS in cancer. Redox Biol 2015;6:334-43.

126. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 2014;6:13.

127. Chen Y, Zhang S, Wang Q, Zhang X. Tumor-recruited M2 macrophages promote gastric and breast cancer metastasis via M2 macrophage-secreted CHI3L1 protein. J Hematol Oncol 2017;10:36.

128. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141:39-51.

129. Vasiliadou I, Holen I. The role of macrophages in bone metastasis. J Bone Oncol 2013;2:158-66.

130. Wyckoff JB, Wang Y, Lin EY, Li JF, Goswami S, et al. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res 2007;67:2649-56.

131. Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med 2001;193:727-40.

132. Monks J, Rosner D, Geske FJ, Lehman L, Hanson L, et al. Epithelial cells as phagocytes: apoptotic epithelial cells are engulfed by mammary alveolar epithelial cells and repress inflammatory mediator release. Cell Death Differ 2005;12:107-14.

133. Monks J, Smith-Steinhart C, Kruk ER, Fadok VA, Henson PM. Epithelial cells remove apoptotic epithelial cells during post-lactation involution of the mouse mammary gland. Biol Reprod 2008;78:586-94.

134. O’Brien J, Martinson H, Durand-Rougely C, Schedin P. Macrophages are crucial for epithelial cell death and adipocyte repopulation during mammary gland involution. Development 2012;139:269-75.

135. Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, et al. Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage-derived VEGFA. Cancer Discov 2015;5:932-43.

136. Holmes S, Downs AM, Fosberry A, Hayes PD, Michalovich D, et al. Sema7A is a potent monocyte stimulator. Scand J Immunol 2002;56:270-5.

137. Hugo HJ, Saunders C, Ramsay RG, Thompson EW. New insights on COX-2 in chronic inflammation driving breast cancer growth and metastasis. J Mammary Gland Biol Neoplasia 2015;20:109-19.

138. Gan L, Qiu Z, Huang J, Li Y, Huang H, et al. Cyclooxygenase-2 in tumor-associated macrophages promotes metastatic potential of breast cancer cells through Akt pathway. Int J Biol Sci 2016;12:1533-43.

139. Esbona K, Yi Y, Saha S, Yu M, Van Doorn RR, et al. The presence of cyclooxygenase 2, tumor-associated macrophages, and collagen alignment as prognostic markers for invasive breast carcinoma patients. Am J Pathol 2018;188:559-73.

140. Ingman WV, Wyckoff J, Gouon-Evans V, Condeelis J, Pollard JW. Macrophages promote collagen fibrillogenesis around terminal end buds of the developing mammary gland. Dev Dyn 2006;235:3222-9.

141. Muthukkaruppan VR, Kubai L, Auerbach R. Tumor-induced neovascularization in the mouse eye. J Natl Cancer Inst 1982;69:699-708.

142. Holmgren L, O’Reilly MS, Folkman J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1995;1:149-53.

143. Parangi S, O’Reilly M, Christofori G, Holmgren L, Grosfeld J, et al. Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc Natl Acad Sci U S A 1996;93:2002-7.

144. Ramirez RA, Lee A, Schedin P, Russell JS, Masso-Welch PA. Alterations in mast cell frequency and relationship to angiogenesis in the rat mammary gland during windows of physiologic tissue remodeling. Dev Dyn 2012;241:890-900.

145. Pepper MS, Baetens D, Mandriota SJ, Di Sanza C, Oikemus S, et al. Regulation of VEGF and VEGF receptor expression in the rodent mammary gland during pregnancy, lactation, and involution. Dev Dyn 2000;218:507-24.

146. Chan TR, Stahl PJ, Li Y, Yu SM. Collagen-gelatin mixtures as wound model, and substrates for VEGF-mimetic peptide binding and endothelial cell activation. Acta Biomater 2015;15:164-72.

147. Bordeleau F, Mason BN, Lollis EM, Mazzola M, Zanotelli MR, et al. Matrix stiffening promotes a tumor vasculature phenotype. Proc Natl Acad Sci U S A 2017;114:492-7.

148. Samani A, Zubovits J, Plewes D. Elastic moduli of normal and pathological human breast tissues: an inversion-technique-based investigation of 169 samples. Phys Med Biol 2007;52:1565-76.

149. Fang M, Yuan J, Peng C, Li Y. Collagen as a double-edged sword in tumor progression. Tumour Biol 2014;35:2871-82.

150. Masson N, Ratcliffe PJ. HIF prolyl and asparaginyl hydroxylases in the biological response to intracellular O(2) levels. J Cell Sci 2003;116:3041-9.

151. Takeda N, Maemura K, Imai Y, Harada T, Kawanami D, et al. Endothelial PAS domain protein 1 gene promotes angiogenesis through the transactivation of both vascular endothelial growth factor and its receptor, Flt-1. Circ Res 2004;95:146-53.

152. Kaidi A, Qualtrough D, Williams AC, Paraskeva C. Direct transcriptional up-regulation of cyclooxygenase-2 by hypoxia-inducible factor (HIF)-1 promotes colorectal tumor cell survival and enhances HIF-1 transcriptional activity during hypoxia. Cancer Res 2006;66:6683-91.

153. Greenhough A, Smartt HJ, Moore AE, Roberts HR, Williams AC, et al. The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 2009;30:377-86.

154. Jones MK, Wang H, Peskar BM, Levin E, Itani RM, et al. Inhibition of angiogenesis by nonsteroidal anti-inflammatory drugs: insight into mechanisms and implications for cancer growth and ulcer healing. Nat Med 1999;5:1418-23.

155. Morote-Garcia JC, Napiwotzky D, Kohler D, Rosenberger P. Endothelial semaphorin 7A promotes neutrophil migration during hypoxia. Proc Natl Acad Sci U S A 2012;109:14146-51.

156. Zhang M, Wang L, Dong M, Li Z, Jin F. Endothelial semaphorin 7A promotes inflammation in seawater aspiration-induced acute lung injury. Int J Mol Sci 2014;15:19650-61.

157. He M, Bian Z. Expression of hypoxia-induced semaphorin 7A correlates with the severity of inflammation and osteoclastogenesis in experimentally induced periapical lesions. Arch Oral Biol 2017;75:114-9.

158. Garcia-Areas R, Libreros S, Amat S, Keating P, Carrio R, et al. Semaphorin7A promotes tumor growth and exerts a pro-angiogenic effect in macrophages of mammary tumor-bearing mice. Front Physiol 2014;5:17.

159. Ghanem RC, Han KY, Rojas J, Ozturk O, Kim DJ, et al. Semaphorin 7A promotes angiogenesis in an experimental corneal neovascularization model. Curr Eye Res 2011;36:989-96.

160. Foskett AM, Ezekiel UR, Trzeciakowski JP, Zawieja DC, Muthuchamy M. Hypoxia and extracellular matrix proteins influence angiogenesis and lymphangiogenesis in mouse embryoid bodies. Front Physiol 2011;2:103.

161. Swartz MA. Immunomodulatory roles of lymphatic vessels in cancer progression. Cancer Immunol Res 2014;2:701-7.

162. Hosono K, Isonaka R, Kawakami T, Narumiya S, Majima M. Signaling of prostaglandin E receptors, EP3 and EP4 facilitates wound healing and lymphangiogenesis with enhanced recruitment of M2 macrophages in mice. PLoS One 2016;11:e0162532.

163. Zeng C, Chen L, Yang Z, Sun S. The close correlation between heparanase and COX-2 expression in lymphangiogenesis of cervical cancer. Med Oncol 2014;31:314.

164. Iwata C, Kano MR, Komuro A, Oka M, Kiyono K, et al. Inhibition of cyclooxygenase-2 suppresses lymph node metastasis via reduction of lymphangiogenesis. Cancer Res 2007;67:10181-9.

165. Lala PK, Nandi P, Majumder M. Roles of prostaglandins in tumor-associated lymphangiogenesis with special reference to breast cancer. Cancer Metastasis Rev 2018;37:369-84.

166. Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res Treat 2010;123:725-31.

167. Burke CA, Phillips R, Berger MF, Li C, Essex MN, et al. Children’s international polyposis (CHIP) study: a randomized, double-blind, placebo-controlled study of celecoxib in children with familial adenomatous polyposis. Clin Exp Gastroenterol 2017;10:177-85.

168. Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, et al. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med 2000;342:1946-52.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Wallace TR, Tarullo SE, Crump LS, Lyons TR. Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms. J Cancer Metastasis Treat 2019;5:9. http://dx.doi.org/10.20517/2394-4722.2019.01

AMA Style

Wallace TR, Tarullo SE, Crump LS, Lyons TR. Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms. Journal of Cancer Metastasis and Treatment. 2019; 5: 9. http://dx.doi.org/10.20517/2394-4722.2019.01

Chicago/Turabian Style

Wallace, Taylor R., Sarah E. Tarullo, Lyndsey S. Crump, Traci R. Lyons. 2019. "Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms" Journal of Cancer Metastasis and Treatment. 5: 9. http://dx.doi.org/10.20517/2394-4722.2019.01

ACS Style

Wallace, TR.; Tarullo SE.; Crump LS.; Lyons TR. Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms. J. Cancer. Metastasis. Treat. 2019, 5, 9. http://dx.doi.org/10.20517/2394-4722.2019.01

About This Article

Special Issue

This article belongs to the Special Issue Breast Cancer Metastasis
© The Author(s) 2019. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
4506
Downloads
1398
Citations
Comments
0
8

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 29 clicks
Like This Article 8 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Journal of Cancer Metastasis and Treatment
ISSN 2454-2857 (Online) 2394-4722 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/