Approximately 60% of metastatic breast cancer patients experience lung or bone metastasis, with a specific predisposition for lung metastasis in basal-like breast cancer (BLBC) or triple-negative breast cancer [
44]. Survival statistics suggest that patients with lung metastasis have a low life expectancy, with a median survival of only 22 months post-treatment [
45]. A significant portion, around 60-70%, of metastatic breast cancer patients who ultimately succumb to the disease are initially diagnosed with lung metastasis [
46]. Despite various available treatment approaches the patient outcomes for BC-LM remains dismally low. The incidence of lung metastasis is notably higher in triple-negative breast cancer (TNBC), reaching up to 40%, compared to only 20% in non-TNBC cases [
47,
48,
49]. Reports suggest that visceral metastasis, specifically pulmonary metastasis, is more common in TNBC patients, while non-TNBC patients tend to develop bone metastasis [
50,
51,
52,
53]. Additionally, luminal A subtype patients tend to avoid lung relapse, while brain metastasis is predominantly observed in those with BLBC and HER2+ breast cancer [
45]. Recent SEER database analysis also confirmed that TNBC, particularly BLBC, is primarily linked to lung metastasis [
54]. BC-LM carries serious clinical implications and consequences, often resulting in a poor prognosis despite treatment strategies such as chemotherapy, targeted therapy, and endocrine therapy based on molecular receptor profiles [
48]. Currently, early diagnosis remains the most effective approach to prevent breast cancer lung metastasis. However, for better patient outcomes with effective therapeutic intervention, when a solitary lung nodule is detected in patients previously treated for breast cancer, it is essential to histologically confirm the diagnosis to differentiate it as recurrent malignancy from primary lung cancer, or benign lung tumors [
49,
55]. Hence, in order to develop more effective diagnostic and therapeutic approaches for BC-LM patients, it is imperative to gain a comprehensive understanding of the underlying mechanisms. In-depth studies of MBC cells and their interactions with the surrounding microenvironment can offer valuable insights into the factors contributing to the existing challenges. Metastasis is an intricate and multi-step process involving various cellular mechanisms such as detachment from the primary tumor, invasion into surrounding tissues, evading the immune system, and altering the local tissue environment [
35,
36]. Large amount of evidence suggests that breast cancer stem cells (BCSCs) are recognized as drivers of metastatic growth [
56,
57]. BCSCs show subtype-specific associations, with studies indicating their enrichment in basal-like breast cancer (BLBC) [
58]. CD44v+ BCSCs in primary tumors are linked to distant metastasis promotion, with their expression enhancing lung metastasis by interacting with lung microenvironment factors [
59]. However, CD44 alone doesn't identify all BCSCs. Enrichment of BCSCs in BC-LM should be further looked into through intricate signaling network regulating their vital properties facilitating invasiveness and poor prognosis [
48]. BCSCs relocating from primary sites to distant microenvironments establish lung niches associated with Notch signaling [
60]. Dysregulated activation of the Notch signaling pathway with Notch1 expression can contribute in unchecked proliferation of BCSCs and affect various aspects of BCSC behavior, including self-renewal, proliferation, apoptosis, and epithelial-mesenchymal transition (EMT) [
61,
62,
63,
64]. Although the precise role of the Notch pathway in primary tumor cell dissemination to the lung is yet unclear, it likely plays a crucial part in adaptation of MBCs to metastatic niches, possibly interacting with other signaling pathways [
48]. The Wnt/β-catenin signaling pathway has a significant role in mammary gland development and breast cancer tumorigenesis; aberrant activation of this pathway through over-expression of β-catenin is associated with worst prognosis in breast cancer, particularly in the triple-negative subtype [
65,
66]. High Wnt/β-catenin signaling is exhibited by BCSCs and is particularly associated with TNBCs advancing EMT and metastasis in breast cancer patients. However, the action of Wnt family members is multifaceted, governed by canonical and non-canonical activation, and can either hinder or drive breast cancer progression and metastasis depending on the specific signaling context [
48]. Additionally, recent research underscores the role of paracrine Hedgehog signaling (Hh) signaling associated with breast cancer growth impacting migration, particularly associated with the poor prognosis of the basal-like phenotype. Studies demonstrate that transcription factors like GLI1 and FOXC1 influence BC-LM through interactions with the CXCL12-CXCR4 axis, controlling angiogenesis, and BCSC properties which are mainly enriched in BLBC [
67,
68,
69]. Dysregulated Hh signaling functions independently as well as through interaction with other signaling pathways in breast cancer. As per a study conducted in hepatocellular carcinoma, the activation of Hh signaling together with TGF- β promoted liver cancer lung metastasis in mouse models, suggesting the same mechanism might be involved in BC-LM which needs to be unveiled [
48,
70]. Thus, co-activation of Hh, Notch, and Wnt pathways in TNBC samples is linked to shorter survival; however, the precise mechanisms of their coordination in breast cancer metastasis are not yet fully understood and investigating their interaction hold promising therapeutic strategies for BC-LM [
71,
72,
73]. Further, the target tissues for disseminated cancer cells (DCCs) engage in diverse interactions with resident stromal cells, immune cells, tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), extra-cellular matrix (ECM), cytokines, chemokines, and other possible factors that are yet-to-be determined [
74,
75]. Studies have indicated that even subtle changes in the composition of leukocytes at distant sites from the primary tumor can influence the development of metastases. CXCR2+ neutrophil subtype has been associated with pro-metastatic effects of mesenchymal-stromal cells [
76]. Recent findings pertaining to breast cancer mouse models illuminate the role of neutrophils in mediating metastatic initiation by modifying the lung microenvironment before metastases. This suggests that immune cells can influence the creation of metastatic niches in BC-LM [
77,
78]. TAMs have also been found to be crucial in BC-LM as the induce EMT through the secretion of CCL18 [
79]. Additionally, clinical studies have linked pulmonary macrophages to promote the survival of breast cancer cells in lung microenvironment by allowing TAMs to interact with VCAM1 receptor which provide survival benefit for MBCs [
79,
80,
81,
82,
83]. Next, the CAFs too, play a significant role in BC-LM as they express Tiam1 and osteopontin that regulate the metastatic process [
84]. Expression of platelet-derived growth factor receptor β (PDGFRβ), a protein associated with CAFs, has been linked to lung metastasis in breast cancer. Key factors influencing the spread of breast cancer to lungs are associated with epithelial to mesenchymal transition, making it a vital and centric process in BC-LM.
1.3.1. Epithelial to Mesenchymal Transition through Invadopodia Formation
EMT is a biological process by which epithelial cells, which are typically immobile and organized in tightly packed structures, acquire characteristics of mesenchymal cells, which are more motile and invasive having the ability to degrade the ECM. Cells in EMT process thus, demonstrate the gain and loss of certain characteristics while switching between two states. It involves complex molecular changes including the down-regulation of epithelial markers (e.g., E-cadherin) and up-regulation of mesenchymal markers (e.g., N-cadherin, vimentin). Additionally, it describes transcription factors like Snail, Slug, and Twist which play a pivotal role in regulating the process [
86]. The regulation of epithelial-mesenchymal transition by TGFβ has been extensively studied and can occur through both SMAD and non-SMAD signaling pathways. Various growth factors, such as EGF, FGF, HGF, and VEGF, stimulate receptor tyrosine kinases (RTKs), initiating multiple signaling cascades that result in the up-regulation of transcription factors associated with EMT. Activation of RTKs or integrins can also activate AKT, leading to increased expression of Snail by inhibiting GSK-3 beta. Furthermore, inflammatory cytokines released by cancer cells itself and also by surrounding immune cells can promote EMT through the induction of Snail and STAT3. Within the tumor microenvironment, the accumulation of HIF-1α drives hypoxic conditions that lead to the expression of TWIST, ultimately inducing EMT [
87]. Besides, molecular drivers, development of invadopodia plays a very crucial role in epithelia-to-mesenchymal transition. Invadopodia are defined as actin based subcellular structures that are described as specialized machinery for extracellular matrix degradation [
88,
89]. Formation of invadopodia is characterized by the development of large protrusions along with branched actin filaments, vesicles and various cytoskeletal components that occupy these protrusions [
90]. Invadopodia influence cell motility through various mechanisms, such as coordinating focal adhesion dynamics, lamellipodia formation, and ECM track generation. They can also act as mechanosensors and exert traction forces on the ECM [
91]. Many studies have illustrated the structural components of invadopodia. However, currently efforts are being made to understand the governing mechanisms of invadopodia formation.
The maturation formation and maturation process of invadopodia involves the recruitment and simultaneous activation of multiple proteases near the cell's periphery, facilitating ECM degradation and release of cytokine. These proteases include zinc-regulated metalloproteases (e.g., MMP2, MMP9, MT1-MMP, ADAM family), cathepsin cysteine proteases, and serine proteases (e.g., seprase and urokinase plasminogen activator) [
92]. Multiple studies have determined that cancer cells stimulated with growth factors such as PDGF, TGFβ, and EGF may trigger the formation of Invadopodia. These stimuli initiate the formation of Invadopodia primarily through their respective signaling cascades with Src and PKC intermediates [
93]. The transition of resting cells to migratory cells takes place through the process of focal adhesion degradation (
Figure 1). Studies have suggested a reciprocal relationship between focal adhesions and Invadopodia, owing to the role of focal adhesion kinases (FAKs) that acts as a negative regulator of Invadopodia. FAK negatively regulate Invadopodia formation by controlling the spatial activation of Src [
94]. Reduction in FAK results in the release of active Src, which enhances the phosphorylation of Invadopodia-related proteins and increase Invadopodia formation. In the Src-transformed cells the initiation of Invadopodia formation occurs near focal adhesions in response to localized production of a lipid called PI3,4-P2. This lipid recruits a protein called Tks5 (hereafter referred as SH3PXD2A), which in turn associates with cortactin (CTTN), a protein crucial for actin regulation. SH3PXD2A is proposed to be the scaffold that recruits cortactin to Invadopodia precursors. SH3PXD2A has the ability to engage with a variety of actin regulatory proteins, including Nck1, Nck2, (N)-WASP, and Grb2. In a similar manner, cortactin also associates with several actin regulatory proteins like (N)-WASP and Arp2/3. It is probable that one or more of these proteins act as intermediaries in the interaction between cortactin and SH3PXD2A. This interaction might be influenced by the phosphorylation state of cortactin, as it can be activated by different kinases such as Src, PAK, and ERK, thereby impacting its interactions with other proteins. Furthermore, cortactin and Tks4 (referred to as SH3PXD2B hereafter) play roles in the advancement of Invadopodia maturation. Cortactin is recognized for its involvement in the secretion of metalloproteases, while SH3PXD2B contributes to the localization or stabilization of MT1-MMPs within Invadopodia, facilitating ECM degradation. Thus, model for Invadopodia maturation demonstrated by Murphy, et.al; states that SH3PXD2A and cortactin cooperate to create invadopodia and release metalloproteases, with SH3PXD2B later aiding in MT1-MMP localization to enable the activation of MMPs and ECM degradation. Therefore, the key players in Invadopodia formation and maturation involve adaptor proteins SH3PXD2B and SH3PXD2A along with cortactin and MMPs [
93].
1.3.2. SH3PXD2B in Cancer Metastasis:
In the recent years, SH3PXD2B a closely related protein to SH3PXD2A has been identified as a critical component of invadopodia in Src-transformed fibroblasts and is implicated in metastasis of melanoma [
95]. A study carried out by Buschman, et.al; have demonstrated that Src-transformed fibroblast cells lacking SH3PXD2B, developed pre-invadopodia structures with essential proteins properly localized for ECM degradation, however due to the lack of SH3PXD2B the degradation did not occur, thus, suggesting its crucial role in functionality of Invadopodia [
96]. This further elucidates that a fully formed and mature invadopodia structure is useless without the presence of SH3PXD2B, and cannot carry out its migratory functions. As discussed, earlier MT1-MMP is recruited by SH3PXD2B and is particularly important for metastasis due to its diverse substrates present in the ECM such as collagens, fibronecting and laminins [
97]. The subcellular localization of MT1-MMP aided by SH3PXD2B is crucial for its functionality in transmembrane domain and cytoplasmic tail as this localization regulates its proteolytic and degradation activities [
98,
99]. This appropriate localization of MT1-MMP thus, contributes to cancer cell growth, studied in 3D ECM environments [
100]. In terms of the functionality of SH3PXD2B in cancer metastasis the recruitment of MT1-MMP results in the activation of MMP2 and MMP9 that degrade the extracellular matrix and facilitate the invasion of process by cancer cells [
96]. Multiples studies have demonstrated that SH3PXD2B plays a significant role in promoting the invasion and metastasis of various cancer types, including colon cancer, breast cancer, and melanoma [
95,
101,
102]. A recent study carried out in oral squamous cell carcinoma highlights that overexpression of crucial proteins associated with invadopodia including SH3PXD2B elevates metastatic capabilities of cancer cells and facilitate tumor progression [
103]. Another recent study investigates the prognostic value of SH3PXD2B in hepatocellular carcinoma (HCC), signifying that high expression of SH3PXD2B is associated with poor overall survival in HCC patients. The findings demonstrate that increased expression of SH3PXD2B in HCC promote tumor growth and metastasis [
104]. Even though the role of SH3PXD2B is not yet explored in BC-LM, these findings suggest that SH3PXD2B could be a potential therapeutic target and curbing its expression may significantly inhibit metastasis.
SH3PXD2B is identified as a scaffold protein responsible for regulating intracellular signaling by bringing regulatory proteins, enzymes, or cytoskeletal structures in close proximity [
105]. The previous studies directed towards the role of SH3PXD2A in cancer metastasis imply that SH3PXD2B may have comparable functions in cancer progression and metastasis due to their evolutionary conservation and structural resemblance. The structural component of SH3PXD2B is characterized by an N-terminal phox homology (PX) domain, four Src homology 3 (SH3) domains, multiple proline-rich motifs (PRMs), and Src phosphorylation sites [
105,
106]. The PX domain's primary function is to join the scaffold protein to the cell membrane through phosphoinositide binding [
96]. SH3 domains act as docking sites for signaling molecules and facilitate protein-protein interactions. Additionally, the proline-rich motifs serve as contact sites for molecules containing the SH3 domains [
105]. SH3PXD2B exhibits two distinct states within cells: a cytoplasmic, inactive state, and a membrane-bound, active state. This transition between states is likely regulated through phosphorylation, although direct evidence for these conformational changes is limited [
96,
107]. This idea is supported by similar self-regulation activity seen in p47phox, a protein that has a slight structural homology with SH3PXD2B with respect to its N-terminal PX domain [
108]. While in cytoplasm, the intramolecular regulatory mechanism in SH3PXD2B includes the binding of SH3 domains to a specific proline-rich motifs within its C-terminal region characterizing its auto-inhibitory state and preventing the PX domain from accessing phosphatidylinositol phosphates, which are important for its function. Consequently, when phosphorylation of C-terminal serine residues that are close to the PRMs takes place this auto-inhibitory state is disrupted and the tandem SH3 domains are exposed, allowing them to bind and interact with their regulatory partners. Simultaneously, the locked PX domain is also freed in order to interact with phospholipids and recruit the MMPs for ECM [
105].
Apart from conferring invasive properties to the cell and orchestrating cell motility, Invadopodia may also have a role in cell-cell communication. SH3PXD2B is reported to bind ADAM15 using its fourth SH3 domain [
109]. The ADAM family proteins are membrane-localized proteases and have the ability to act as sheddases. They are involved in the activation of growth factors or ligands by cleaving their inactive membrane-anchored forms and release their active forms. This shedding process has been demonstrated for various molecules, including insulin-like growth factor-binding protein (IGF-BP), Delta-like ligand 1 (DLL1), E-cadherin, amphiregulin, heparin-binding EGF-like growth factor (HB-EGF), transforming growth factor alpha (TGFα), EGF, and tumor necrosis factor alpha (TNFα). After cleavage by ADAMs, the released ligands exert their effects on the same, adjacent, or distant cells, facilitating communication between the "signal sender" cell and "receiver" cells [
105]. Even though the role of SH3PXD2B has been critically highlighted in cancer metastasis, certain studies suggest its contradictory role. It has been recognized with a novel function in negatively regulating the EMT processes in few cancer models. A study conducted in colon cancer found that SH3PXD2B found to play a part in preventing EMT-like changes in colon cancer cells and when it was knocked down from these cells, they began to display characteristics associated with mesenchymal cells facilitating EMT [
110]. However, the exact mechanism and factors associated with the contradictory functions of SH3PXD2B in cancer metastasis is not completely clear and deeper research is needed to elucidate its appropriate functions.
Understanding the contradictory role of SH3PXD2B in cancer metastasis and deciphering its functionality in breast cancer-lung metastasis model requires a multidisciplinary approach that combines cancer systems biology and computational structure biology. Through this approach our study aims at integrating multiomics data from breast cancer patients and breast cancer cell lines with different metastatic outcomes to discern patterns linked to SH3PXD2B expression and its correlation with metastasis. Additionally, it involves construction of comprehensive and inter-regulatory protein-protein networks to unveil potential functional modules and pathways associated with SH3PXD2B. Finally, through the use of computational structure biology platform we seek to dive deeper into the structural aspects SH3PXD2B by eliminating disordered regions and revealing potential target sites for therapeutic interventions.