Biological implications of decoding the extracellular matrix of vulva cancer
- Authors:
- Published online on: December 6, 2024 https://doi.org/10.3892/or.2024.8852
- Article Number: 19
-
Copyright: © Islam et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Vulvar cancer is the fourth most prevalent gynecological cancer globally, representing 5% of lower genital tract tumors and ranking below uterine corpus, ovarian and cervical cancer (1–3). Key contributors to vulvar cancer development include age, human papillomavirus (HPV) infection, smoking, inflammatory vulvar conditions, prior pelvic radiation, immunodeficiency and anogenital warts (4–6). Among the different histological types of vulvar cancer, vulvar squamous cell carcinoma (VSCC) is the most common type (95%), followed by melanoma, sarcoma and basal cell carcinoma (2,3). VSCC has traditionally been regarded as a disease of postmenopausal women, although the mean age of incidence has fallen in recent years worldwide owing to the increase in HPV infections (1). Nonetheless, the age-specific incidence ranges from 0.4 per 100,000 in younger women to 20 per 100,000 in women >70 years old (7).
VSCC manifests in two types with different pathways: The basaloid and/or warty type often associated with HPV (HPV subtypes 16, 18, 31 and 33) which is predisposed from usual vulvar intraepithelial neoplasia (uVIN), and the second type linked to chronic vulvar dermatoses and differentiated vulvar intraepithelial neoplasia (dVIN) (8). Common TP53 mutations are observed in the second type and are mostly independent of HPV (8). Although there are survival differences in these two types of VSCC, treatment is predominantly surgery with or without radio-chemotherapy. Immunotherapy has reformed the therapeutic paradigms of multiple malignancies, but its impact is limited in VSCC (1). In addition, treatment of unresectable/metastatic disease often leads to frequent comorbidities, particularly in elderly and frail women, highlighting the need for innovative and more effective treatment approaches.
Carcinomas comprise both malignant and non-malignant cells, including fibroblasts, immune cells, vascular cells and neuronal cells (9). Non-malignant cells actively shape the tumor microenvironment (TME) by secreting cytokines, chemokines, growth factors and extracellular matrix (ECM) proteins (9). Under normal physiological conditions, resident cells maintain tissue balance through ECM deposition, degradation and remodeling (10). However, carcinogenesis disrupts ECM remodeling, creating a dysfunctional ECM conducive to a supportive TME that promotes cancer growth, invasion and metastasis (11). While specific research on ECM proteins in VSCC is limited, studies are starting to explore the role of ECM remodeling in VSCC invasion and metastasis.
The present review aims to provide an overview on the role of ECM during VSCC metastasis and the current understanding of the role of ECM in regulating VSCC dissemination. The present study also explored the therapeutic potential of targeting ECM in other types of squamous-epithelial cancers, and the potential prognostic and predictive biomarkers, discussing their impact on developing more efficacious antitumor therapies.
Overview of ECM components
The ECM is commonly defined as the non-malignant, non-cellular component of tissue that provides essential biochemical and structural support to its cellular constituents (12,13). Emerging research suggests that the ECM is not merely an intercellular filler but a physiologically active component of living tissue, playing crucial roles in cell-cell communication, adhesion and proliferation (14,15). Resident fibroblasts are responsible for creating and arranging ECM components according to the specific needs of the tissue (14,16). Major components of the ECM, including collagen, laminin, elastin and proteoglycans, exhibit distinct physical and biochemical properties. A detailed overview of ECM components is shown in Table I.
ECM remodeling in tumorigenesis
Dysregulation of the ECM is a critical factor in cancer development and progression, influencing various key mechanisms. One such mechanism is cellular signaling, where abnormalities in the ECM contribute to uncontrolled cell growth, survival and proliferation, all fundamental cancer hallmarks (14). Moreover, the ECM plays a critical role in cell adhesion and migration, facilitated by proteins such as integrins and cadherins. Disruption of these processes promotes the invasion of cancer cells into surrounding tissues (Fig. 1A) (10). The ECM also affects the immune response within TME, contributing to immune evasion by cancer cells (14). It has been shown that ECM components impede and educate immune cell types such as natural killer cells, macrophages and tumor-infiltrating lymphocytes, specifically CD8+ T cells, within the TME and evade the antitumor immune response (14,17,18). Components such as cross-linked collagen, fibronectin, laminin, periostin, osteopontin, integrins and matrix metalloproteinases (MMPs) can erect physical barriers, hindering immune cell movement and impairing the ability of immune cells such as cytotoxic CD8+ T-cells to target and eliminate cancer cells (Fig. 1B) (19,20).
Understanding the intricate interplay between tumorigenesis and the ECM is essential for developing targeted therapeutic approaches. Fig. 1C illustrates the significant involvement of ECM components in various cancer hallmarks. Over time, intensive research has been dedicated to exploring interventions targeting ECM components or disrupting ECM-associated signaling pathways in tumors originating from squamous epithelia. These endeavors have yielded valuable scientific insights, which are outlined in Table II, and offer promising directions for advancing SCCs treatment (21–102).
Navigating the matrix: Understanding ECM dynamics in vulvar cancer
The ECM in VSCC is a dynamic and influential factor in tumor development (11). This intricate network of proteins, including collagens, laminin, osteopontin, dystroglycan, integrin, CD44 and MMPs, significantly contributes to the progression of VSCC (11). Fig. 1D provides a concise summary of the roles of specific ECM proteins in VSCC and their effects on the hallmarks of cancer. These hallmarks include invasion and metastasis, resistance to cell death, promotion of inflammation, facilitation of replicative immortality and evasion of immune destruction. Thus, understanding the ECM landscape in VSCC can provide valuable insights into the molecular mechanisms underlying this type of cancer.
In order to decipher key ECM proteins involved in VSCC progression, a thorough literature search was performed in the present study. The following MeSH terms were used in PubMed (https://pubmed.ncbi.nlm.nih.gov/), Scopus (https://www.scopus.com/home.uri) and Web of Science (https://mjl.clarivate.com/home) to select literature describing ECM and VSCC tumor progression and development: (‘genital neoplasms, female’[MeSH Terms] OR ‘female genital neoplasm*’[All Fields] OR ‘Gynaecologic neoplasm*’[All Fields] OR ‘Vulvar Neoplasms’[MeSH Terms] OR ‘vulvar neoplasm*’[All Fields] OR ‘vulva neoplasm*’[All Fields] OR ‘cancer of vagina*’[All Fields] OR ‘cancer of vulva*’[All Fields] OR ‘vulva squamous cell carcinoma*’[All Fields] OR ‘vaginal neoplasm*’[All Fields] OR ‘vagina cancer*’[All Fields]) AND (‘Extracellular Matrix’[MeSH Terms] OR ‘extracellular matrix*’[All Fields] OR ‘extracellular matrix protein*’[All Fields]) AND 1980/01/01:2023/12/31 [date-publication]. In the following section, each of the ECM proteins implicated in VSCC progression are described in detail. Table III (103–115) summarizes the key findings regarding ECM proteins in VSCC. This table provides a concise overview of the significant associations, functional implications and clinical relevance of ECM proteins identified in VSCC.
Collagens
Collagens, which are the predominant component of the ECM, are widely distributed across various types of tissue. With 28 different types, the collagen superfamily forms fibers, networks and filaments within the ECM, interacting with mesenchymal-origin cells through various receptor families to regulate their proliferation, migration and differentiation (116). While studies have extensively investigated the role of collagens in driving cancer invasion and metastasis, their specific involvement in VSCC remains understudied. Recent research utilizing Second Harmonic Generation imaging has analyzed collagen parameters (quantity, uniformity and organization) in VSCC, revealing associations with lymph node metastasis (103). Additionally, based on collagen organization, two morphologic variants have been identified in VSCC: An indolent type growing as ‘sheets of cells’ with a pushing border in lymphoplasmacytic stroma, and an aggressive variant growing as ‘single tumor cells’ with a finger-like border in fibromyxoid stroma (104). Proteomic analyses have further demonstrated that the aggressive variants are associated with higher rates of lymph node metastasis and tumor recurrences. Consistent with these findings, previous studies have shown a significant correlation between the ‘sheet of cells’ morphology, HPV infection and improved survival rate (8). Collectively, these findings suggest that morphological variants of collagen fibers in the TME may serve as prognostic indicators for aggressive VSCC. This highlights the potential significance of collagen parameters in understanding VSCC aggressiveness and may offer insights into therapeutic strategies targeting the tumor microenvironment.
Laminins
The laminin family, comprising of ~20 glycoproteins, forms a cross-linked web intertwined with the type IV collagen network in basement membranes. Laminins are heterotrimers composed of three polypeptide chains (α, β, γ) and play crucial roles in early embryonic development, organogenesis and various cell type-specific functions such as adhesion, differentiation, migration, phenotype maintenance and resistance to apoptosis (15). However, in tumors of the lower female genital tract, laminin expression becomes dysregulated (117). Prior research has emphasized that elevated expression of the γ2 chain of laminin-5 (LAMC2) is linked to patient survival in VSCC. Notably, intracytoplasmic expression of the γ2 chain along the invasive tumor front correlates with short-term survival. Larger tumors tend to exhibit increased γ2 chain expression, although no significant correlation has been observed with tumor staging. These findings suggest that heightened expression of the γ2 chain may be involved in the initiation of VSCC tumorigenesis rather than progression (115). However, further investigations are warranted to elucidate the dynamics of LAMC2 in VSCC tumorigenesis.
Osteopontin
Matricellular proteins form a diverse family of non-structural matrix glycoproteins, including thrombospondins, secreted acidic protein and rich in cysteine, tenascins, fibulins, osteopontin, cartilage oligomeric matrix protein and CNN family proteins such as periostin and R-spondins (116). Among these, osteopontin (OPN), initially identified as a bone matrix protein, is now recognized as a cytokine produced by activated T cells and transformed cells. It is highly inducible as it is expressed and secreted by both tumor cells and cells in the stroma (105). In VSCC, OPN expression was investigated across various stages of vulvar lesions, including VSCC, VIN, vulvar lichen sclerosus (VLS) and normal vulvar tissue samples. Proteomic analysis revealed a gradual increase in OPN expression from VIN to VLS, with the highest expression observed in VSCC tumor tissue samples. Additionally, OPN expression was found to be associated with the pathological stage, suggesting its potential role in VSCC tumor progression through neoplastic transformation (105). Collectively, this observation suggested that OPN could be a predictive biomarker for the early detection of VSCC, and further studies are required to understand VSCC pathogenesis through OPN.
Dystroglycan
Dystroglycan, a transmembrane glycoprotein, serves as a crucial link between the ECM and the intracellular cytoskeleton, thereby providing structural integrity. Comprising α and β components, dystroglycan facilitates cell adhesion to the ECM and plays a pivotal role in regulating cytoskeletal organization (118,119). Dysregulation of dystroglycan is a common occurrence observed in various human epithelial cancers, suggesting its potential involvement in tumor development (118,119). Notably, previous research has revealed disrupted expression levels of α-dystroglycan in conditions such as VLS, squamous cell hyperplasia, VIN and invasive VSCC. Specifically, decreased expression of α-dystroglycan has been observed across preneoplastic lesions of VSCC, and this downregulation is associated with advanced stages of VSCC. These findings suggest that α-dystroglycan may play a significant role in maintaining cytoskeletal dynamics, and its reduced expression could promote VSCC progression (106).
Integrins β1
Integrins, heterodimeric receptors comprising α and β subunits, are frequently dysregulated in skin cancers (107). Serving as bridging molecules, integrins connect ECMs with the cell cytoskeleton, governing cell adhesion and motility. The intracellular tail of integrin β1 associates with proteins such as talin, α-actinin and vinculin, linking it to the actin cytoskeleton and regulating cell motility, keratinocyte wound healing and the collective movement of tumor cells (115). Increased expression of various integrin (ITG) family proteins, including α2, α3, α5, α6 and β1 has been observed in VSCC. Among these, β1 (ITGB1) plays a pivotal role in mediating cell adhesion, migration and invasion (107). Knockdown experiments targeting β1 result in significant alterations in VSCC tumor morphology compared with control tumors. Specifically, β1 knockdown leads to a more encapsulated and less invasive tumor phenotype, indicating the crucial involvement of integrin β1 in VSCC invasiveness and disease progression (107). The present study underscores the significance of β1 integrin in VSCC tumor advancement and suggests potential therapeutic avenues for intervention.
Hyaluronic acid receptor CD44
Hyaluronic acid receptor CD44 is a surface-expressed glycoprotein that facilitates interactions with a spectrum of molecules, including collagen, fibronectin, OPN, MMPs and growth factors (70,71). CD44 plays a pivotal role in cell adhesion, interactions, migration and metastasis. CD44 isoforms bolster malignant cell affinity to ECM ligands, thereby fostering tumor dissemination (70,71). A previous study has demonstrated a significant association between CD44 variants, particularly CD44v3 and CD44v6 and VSCC tumor progression, as well as adverse patient outcomes (108). Elevated CD44 expression correlates with poor tumor differentiation, positive lymph node involvement, advanced-stage VSCC and diminished survival rates, indicating its potential as a prognostic marker (108).
MMPs
MMPs are a family of calcium-dependent, zinc-containing endopeptidases that target various molecules, including matrix components, growth factors, cytokines and signaling molecules. Synthesized as zymogens, MMPs are secreted after cleavage of their propeptide form. Invasion and metastasis of malignant cells involve the degradation of the stromal matrix, mediated by specific MMPs (120).
MMP2
MMP2 plays a pivotal role in degrading crucial components of basement membranes such as type IV collagen and fibronectin, facilitating the invasion of tumor cells into stromal and vascular regions (25,110). Overexpression of MMP2 has been observed across various disease stages of VSCC, including VIN (grades-I, II, III) and VLS and this heightened expression is significantly associated with the invasiveness of VSCC (110). Morphologically, MMP2 manifests as cytoplasmic granular or diffuse staining in stromal cells. However, in cases of invasive VSCC, MMP2-positive cells are notably observed in the stroma adjacent to neoplastic islands or infiltrating groups of tumor cells (110). These highly MMP2-expressing tumor cells secrete factors that contribute to the aggressiveness of VSCC, including invasion and metastasis, suggesting potential therapeutic approaches targeting MMP2.
MMP12
MMP12, recognized for its ability to degrade elastin and various substrates such as type IV collagen, fibronectin and laminin, plays a multifaceted role in cancer progression, particularly in VSCC. It contributes to limiting tumor growth by converting plasminogen into angiostatin, which inhibits endothelial cell proliferation and angiogenesis, essential processes for tumor vascularization (111). While typically associated with macrophages, MMP12 is also expressed by transformed epithelial cells in VSCC, with its expression level correlating with tumor dedifferentiation and histological aggressiveness (112). In a study involving 33 VSCC samples, MMP12 mRNA was prevalent, and higher expression in cancer cells was associated with more aggressive and poorly differentiated tumors (112). Notably, macrophage-derived MMP12 was more abundant in well-differentiated grade I tumors compared with grade III tumors (112). These findings suggest MMP12 potential role in modulating VSCC histology and influencing treatment strategies. However, further research is warranted to fully understand MMP12 implications in VSCC and its therapeutic relevance.
MMP13
MMP13, recognized for its ability to cleave fibrillar collagens and various stromal matrix components, stands out as a versatile proteolytic tool crucial for tumor cell invasion (16,116,120,121). A previous study has highlighted the abundant expression of MMP13 in VSCC tumor tissues, often correlating with lymph node metastasis (109). Moreover, in vitro investigations have revealed a significant increase in MMP13 levels in VSCC cell lines compared with control epithelial cells (109). These findings underscore MMP13 specific expression by malignantly transformed squamous epithelial cells, including VSCC cells, suggesting its potential as a marker for their invasive potential (109).
Elucidating molecular synergies: Cancer-associated fibroblasts (CAFs) and ECM in VSCC progression
Fibroblasts are the main architects of the ECM, orchestrating changes that play a major role in tumor progression, inflammation, therapy resistance and immunosuppression (9,122). CAFs are perhaps the cells most proficient at remodeling the ECM as they deposit collagens and release various growth factors, chemokines, cytokines and MMPs (Fig. 1A). Significantly altered gene pathways in CAFs include regulation of substrate adhesion, tissue remodeling, cell migration, secretion, growth regulation and angiogenesis (123). Although, at present, VSCC-specific subpopulations of CAFs have not been reported, single-cell RNA sequencing data suggests that various CAF subpopulations exist in the TME of other types of SCCs (124,125).
The most commonly identified CAF subpopulations are those involved in ECM remodeling [myofibroblast CAFs (myCAFs)] and immunomodulatory cancer-associated fibroblasts] (126). MyCAFs have been reported to be involved in tissue remodeling and often express ACTA2 gene (126). Inflammatory CAFs often exhibit increased IL-6 signaling in various tumor types (126–129). This makes them potential targets for anti-IL-6 therapies, such as siltuximab and tocilizumab (130). Moreover, research has shown that immunoregulatory CAFs can activate JAK-STAT signaling in cancer cells (129), suggesting that inhibition of this pathway may be a promising therapeutic strategy.
Mounting evidence suggests that CAFs also play a key role in therapy-resistance in SCCs, particularly in head and neck SCC (HNSCC) and esophageal SCC (131–133). It has been previously reported that chemotherapy promotes CAF survival and alters exosome biogenesis, which leads to malignant characteristics in HNSCC (134). Notably, CAFs also respond to radiotherapy by upregulating transforming growth factor β1 (TGFβ1) expression via IL-8/NF-κB signaling in HNSCC, which increases the rate of invasive growth. This signaling can be inhibited by using Tranilast, a known TGFβ1 inhibitor (135). A subset of CAFs have also been reported to be involved in TGFβ1 dependent PD-1+/TIM-3+ exhaustion of CD8 T cells in HNSCC, leading to exclusion of T cells, thereby negatively impacting immunotherapy response (124,136). Previously, we have shown that CAFs play a decisive role in VSCC invasion and that VSCC cells exhibit a fibroblast-dependent tumorigenic potential (137). However, there exists a significant heterogeneity with CAFs showing both tumor-promoting and tumor-inhibiting CAFs (9,122). Gaining a more profound insight into the molecular and phenotypic variations among CAF populations in VSCC may contribute to overcoming challenges related to therapy resistance and the targeting of cancer cells.
Decoding therapeutic prospects: ECM proteins in VSCC
The therapeutic potential of ECM proteins in treating VSCC holds promise for pioneering treatment strategies. Identifying specific ECM proteins involved in VSCC tumorigenesis will unveil potential therapeutic targets. While the pathways linked with ECM proteins in VSCC are not fully understood, emerging bioinformatic platforms (138) provide opportunities to pinpoint key pathways and potential targeted therapies for this relatively underexplored cancer. Notably, these proteins, irrespective of ECM organization, are implicated in modulating immune systems, as depicted in Fig. 2.
Despite the absence of reports on ECM proteins regulating immune compartments in VSCC, insights from other epithelial-origin tumors in The Cancer Genome Atlas suggests that investigating ECM in VSCC may be beneficial. Bioinformatics platforms such as TIMER2 (http://timer.cistrome.org/) (139) confirm that higher expression levels of ECM-related genes are associated with reduced CD8+ T cell infiltration within the TME, fostering immune evasion and tumor progression across various epithelial cancers (140–143) (Figs. 3 and 4). Consequently, over the last few decades, several ECM inhibitors have been developed and successfully employed across different SCCs (Table IV) (144–158). Therefore, understanding the intricate relationship between ECM components in the context of VSCC is essential for devising innovative therapeutic strategies tailored to this patient population. Such endeavors hold the potential to pave the way for the emergence of novel therapeutics.
Exploring future avenues: Research directions in VSCC and ECM interactions
The present study thoroughly investigated the intricate landscape of the ECM in VSCC, examining the roles of various components such as collagens, laminin, osteopontin, dystroglycan, integrin, CD44 and MMPs in VSCC progression. Furthermore, the present study highlights the evolving understanding of ECM, going beyond its structural role to actively influence adhesion, proliferation and cell communication. Attention is provided to collagen types, emphasizing their structural significance and their correlation with changes in collagen levels and lymph node metastasis in VSCC.
Conclusion
The present study acknowledges the rising incidence of vulvar cancer and the limitations of current treatment approaches, particularly in incurable or metastatic cases. It advocates for the exploration of novel therapeutic pathways and targets based on the detailed analysis of ECM components in VSCC. Recognizing the complex interactions within the TME underscores the necessity for innovative and targeted therapeutic approaches to enhance outcomes for individuals affected by VSCC. Further research and clinical exploration of these ECM targets could pave the way for more effective therapeutic strategies in the management of VSCC.
Acknowledgements
The results shown here are in part based upon data generated by the TCGA Research Network: https://www.cancer.gov/tcga.
Funding
This work was supported by the Research Council of Norway through its Centers of Excellence funding scheme (grant no. 22325).
Availability of data and materials
Not applicable.
Authors' contributions
EI and KCD conducted the literature review and drafted the manuscript. RM, KO, SI and HND contributed to the writing of the manuscript and made corrections. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Olawaiye AB, Cuello MA and Rogers LJ: Cancer of the vulva: 2021 update. Int J Gynaecol Obstet. 155 (Suppl 1):7–18. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rogers LJ and Cuello MA: Cancer of the vulva. Int J Gynaecol Obstet. 143 (Suppl 2):S4–S13. 2018. View Article : Google Scholar | |
Alkatout I, Schubert M, Garbrecht N, Weigel MT, Jonat W, Mundhenke C and Günther V: Vulvar cancer: Epidemiology, clinical presentation, and management options. Int J Womens Health. 7:305–313. 2015. View Article : Google Scholar : PubMed/NCBI | |
Capria A, Tahir N and Fatehi M: Vulva Cancer. StatPearls. StatPearls Publishing; Treasure Island, FL: 2024, PubMed/NCBI | |
Bucchi L, Pizzato M, Rosso S and Ferretti S: New insights into the epidemiology of vulvar cancer: Systematic literature review for an update of incidence and risk factors. Cancers (Basel). 14:3892022. View Article : Google Scholar : PubMed/NCBI | |
Brinton LA, Thistle JE, Liao LM and Trabert B: Epidemiology of vulvar neoplasia in the NIH-AARP Study. Gynecol Oncol. 145:298–304. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gadducci A, Tana R, Barsotti C, Guerrieri ME and Genazzani AR: Clinico-pathological and biological prognostic variables in squamous cell carcinoma of the vulva. Crit Rev Oncol Hematol. 83:71–83. 2012. View Article : Google Scholar : PubMed/NCBI | |
Dongre HN, Elnour R, Tornaas S, Fromreide S, Thomsen LCV, Kolseth IBM, Nginamau ES, Johannessen AC, Vintermyr OK, Costea DE and Bjørge L: TP53 mutation and human papilloma virus status as independent prognostic factors in a Norwegian cohort of vulva squamous cell carcinoma. Acta Obstet Gynecol Scand. 103:165–175. 2024. View Article : Google Scholar : PubMed/NCBI | |
Dongre H and Costea DE: Tumor-Fibroblast Interactions in CarcinomasBiomarkers of the Tumor Microenvironment. Springer; New York, NY: pp. 109–124. 2022, View Article : Google Scholar | |
Dzobo K and Dandara C: The Extracellular Matrix: Its composition, function, remodeling, and role in tumorigenesis. Biomimetics (Basel). 8:1462023. View Article : Google Scholar : PubMed/NCBI | |
Pappa KI, Jacob-Hirsch J, Vlachos GD, Christodoulou I, Partsinevelos G, Amariglio N, Markaki S, Antsaklis A and Anagnou NP: Expression profiling of vulvar carcinoma: clues for deranged extracellular matrix remodeling and effects on multiple signaling pathways combined with discrete patient subsets. Transl Oncol. 4:301–313. 2011. View Article : Google Scholar : PubMed/NCBI | |
Sleeboom JJF, van Tienderen GS, Schenke-Layland K, van der Laan LJW, Khalil AA and Verstegen MMA: The extracellular matrix as hallmark of cancer and metastasis: From biomechanics to therapeutic targets. Sci Transl Med. 16:eadg38402024. View Article : Google Scholar : PubMed/NCBI | |
Mukherjee A and Bravo-Cordero JJ: Regulation of dormancy during tumor dissemination: the role of the ECM. Cancer Metastasis Rev. 42:99–112. 2023. View Article : Google Scholar : PubMed/NCBI | |
Popova NV and Jücker M: The functional role of extracellular matrix proteins in cancer. Cancers (Basel). 14:2382022. View Article : Google Scholar : PubMed/NCBI | |
Walker C, Mojares E and Del Río Hernández A: Role of extracellular matrix in development and cancer progression. Int J Mol Sci. 19:30282018. View Article : Google Scholar : PubMed/NCBI | |
Yue B: Biology of the extracellular matrix: An overview. J Glaucoma. 23 (8 Suppl 1):S20–S23. 2014. View Article : Google Scholar : PubMed/NCBI | |
Condic M, Rohr A, Riemann S, Staerk C, Ayub TH, Doeser A, Zillinger T, Merkelbach-Bruse S, Buettner R, Barchet W, et al: Immune profiling of vulvar squamous cell cancer discovers a macrophage-rich subtype associated with poor prognosis. Cancer Res Commun. 4:861–875. 2024. View Article : Google Scholar : PubMed/NCBI | |
van Esch EM, van Poelgeest MI, Trimbos JB, Fleuren GJ, Jordanova ES and van der Burg SH: Intraepithelial macrophage infiltration is related to a high number of regulatory T cells and promotes a progressive course of HPV-induced vulvar neoplasia. Int J Cancer. 136:E85–E94. 2015. View Article : Google Scholar : PubMed/NCBI | |
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI and Schillaci R: Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol. 13:11354562023. View Article : Google Scholar : PubMed/NCBI | |
Li L, Wei JR, Dong J, Lin QG, Tang H, Jia YX, Tan W, Chen QY, Zeng TT, Xing S, et al: Laminin γ2-mediating T cell exclusion attenuates response to anti-PD-1 therapy. Sci Adv. 7:eabc83462021. View Article : Google Scholar : PubMed/NCBI | |
Zhang W, Huang X, Huang R, Zhu H, Ye P, Lin X, Zhang S, Wu M and Jiang F: MMP1 overexpression promotes cancer progression and associates with poor outcome in head and neck carcinoma. Comput Math Methods Med. 2022:30583422022.PubMed/NCBI | |
Liu M, Hu Y, Zhang MF, Luo KJ, Xie XY, Wen J, Fu JH and Yang H: MMP1 promotes tumor growth and metastasis in esophageal squamous cell carcinoma. Cancer Lett. 377:97–104. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kurnia I, Rauf S, Hatta M, Arifuddin S, Hidayat YM, Natzir R, Kaelan C, Bukhari A, Pelupessy NU and Patelonggi IJ: Molecular Patho-mechanisms of cervical cancer (MMP1). Ann Med Surg (Lond). 77:1034152022.PubMed/NCBI | |
Han L, Sheng B, Zeng Q, Yao W and Jiang Q: Correlation between MMP2 expression in lung cancer tissues and clinical parameters: A retrospective clinical analysis. BMC Pulm Med. 20:2832020. View Article : Google Scholar : PubMed/NCBI | |
Samantaray S, Sharma R, Chattopadhyaya TK, Gupta SD and Ralhan R: Increased expression of MMP-2 and MMP-9 in esophageal squamous cell carcinoma. J Cancer Res Clin Oncol. 130:37–44. 2004. View Article : Google Scholar : PubMed/NCBI | |
Azevedo Martins JM, Rabelo-Santos SH, do Amaral Westin MC and Zeferino LC: Tumoral and stromal expression of MMP-2, MMP-9, MMP-14, TIMP-1, TIMP-2, and VEGF-A in cervical cancer patient survival: A competing risk analysis. BMC Cancer. 20:6602020. View Article : Google Scholar : PubMed/NCBI | |
Li H, Yang F, Chai L, Zhang L, Li S, Xu Z and Kong L: CCAAT/Enhancer Binding Protein β-Mediated MMP3 upregulation promotes esophageal squamous cell cancer invasion in vitro and is associated with metastasis in human patients. Genet Test Mol Biomarkers. 23:304–309. 2019. View Article : Google Scholar : PubMed/NCBI | |
Gkouveris I, Nikitakis NG, Aseervatham J, Rao N and Ogbureke KUE: Matrix metalloproteinases in head and neck cancer: Current perspectives. Metalloproteinases Med. 4:47–61. 2017. View Article : Google Scholar | |
Shao L, Wang X, Liu W, Zhang C, Ma W, Yu X and Han J: The role and function of secretory protein MMP3 in cervical cancer. researchsquare. https://doi.org/10.21203/rs.3.rs-2449297/v1 | |
Liu D, Nakano J, Ishikawa S, Yokomise H, Ueno M, Kadota K, Urushihara M and Huang CL: Overexpression of matrix metalloproteinase-7 (MMP-7) correlates with tumor proliferation, and a poor prognosis in non-small cell lung cancer. Lung Cancer. 58:384–391. 2007. View Article : Google Scholar : PubMed/NCBI | |
Chuang HC, Su CY, Huang HY, Huang CC, Chien CY, Du YY and Chuang JH: Active matrix metalloproteinase-7 is associated with invasion in buccal squamous cell carcinoma. Mod Pathol. 21:1444–1450. 2008. View Article : Google Scholar : PubMed/NCBI | |
Zhu L, Zheng X, Du Y, Xing Y, Xu K and Cui L: Matrix metalloproteinase-7 may serve as a novel biomarker for cervical cancer. Onco Targets Ther. 11:4207–4220. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Ma J, Guo Q, Duan F, Tang F, Zheng P, Zhao Z and Lu G: Overexpression of MMP-2 and MMP-9 in esophageal squamous cell carcinoma. Dis Esophagus. 22:664–667. 2009. View Article : Google Scholar : PubMed/NCBI | |
Tsukamoto S, Koma YI, Kitamura Y, Tanigawa K, Azumi Y, Miyako S, Urakami S, Hosono M, Kodama T, Nishio M, et al: Matrix metalloproteinase 9 induced in esophageal squamous cell carcinoma cells via close contact with tumor-associated macrophages contributes to cancer progression and poor prognosis. Cancers (Basel). 15:29872023. View Article : Google Scholar : PubMed/NCBI | |
Deraz EM, Kudo Y, Yoshida M, Obayashi M, Tsunematsu T, Tani H, Siriwardena SB, Keikhaee MR, Qi G, Iizuka S, et al: MMP-10/stromelysin-2 promotes invasion of head and neck cancer. PLoS One. 6:e254382011. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Qin YR, Bi J, Guo A, Fu L and Guan XY: Overexpression of matrix metalloproteinase 10 is associated with poor survival in patients with early stage of esophageal squamous cell carcinoma. Dis Esophagus. 25:656–663. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhang G, Miyake M, Lawton A, Goodison S and Rosser CJ: Matrix metalloproteinase-10 promotes tumor progression through regulation of angiogenic and apoptotic pathways in cervical tumors. BMC Cancer. 14:3102014. View Article : Google Scholar : PubMed/NCBI | |
Ma B, Ran R, Liao HY and Zhang HH: The paradoxical role of matrix metalloproteinase-11 in cancer. Biomed Pharmacother. 141:1118992021. View Article : Google Scholar : PubMed/NCBI | |
Hsin CH, Chou YE, Yang SF, Su SC, Chuang YT, Lin SH and Lin CW: MMP-11 promoted the oral cancer migration and Fak/Src activation. Oncotarget. 8:32783–32793. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lv FZ, Wang JL, Wu Y, Chen HF and Shen XY: Knockdown of MMP12 inhibits the growth and invasion of lung adenocarcinoma cells. Int J Immunopathol Pharmacol. 28:77–84. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kim JM, Kim HJ, Koo BS, Rha KS and Yoon YH: Expression of matrix metalloproteinase-12 is correlated with extracapsular spread of tumor from nodes with metastasis in head and neck squamous cell carcinoma. Eur Arch Otorhinolaryngol. 270:1137–1142. 2013. View Article : Google Scholar : PubMed/NCBI | |
Han F, Zhang S, Zhang L and Hao Q: The overexpression and predictive significance of MMP-12 in esophageal squamous cell carcinoma. Pathol Res Pract. 213:1519–1522. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kudo Y, Iizuka S, Yoshida M, Tsunematsu T, Kondo T, Subarnbhesaj A, Deraz EM, Siriwardena SB, Tahara H, Ishimaru N, et al: Matrix metalloproteinase-13 (MMP-13) directly and indirectly promotes tumor angiogenesis. J Biol Chem. 287:38716–38728. 2012. View Article : Google Scholar : PubMed/NCBI | |
Jiao XL, Chen D, Wang JG and Zhang KJ: Clinical significance of serum matrix metalloproteinase-13 levels in patients with esophageal squamous cell carcinoma (ESCC). Eur Rev Med Pharmacol Sci. 18:509–515. 2014.PubMed/NCBI | |
Zheng Cl, Lu Q, Zhang N, Jing PY, Zhang JP, Wang WP and Li GZ: Comprehensive analysis of the immune and prognostic implication of MMP14 in lung cancer. Dis Markers. 2021:59175062021. View Article : Google Scholar : PubMed/NCBI | |
Kurahara S, Shinohara M, Ikebe T, Nakamura S, Beppu M, Hiraki A, Takeuchi H and Shirasuna K: Expression of MMPS, MT-MMP, and TIMPs in squamous cell carcinoma of the oral cavity: Correlations with tumor invasion and metastasis. Head Neck. 21:627–638. 1999. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Zhang Y, Zhang Y, Liu W and Wang J: Cryptotanshinone inhibits lung cancer invasion via microRNA-133a/matrix metalloproteinase 14 regulation. Oncol Lett. 18:2554–2559. 2019.PubMed/NCBI | |
Chen N, Zhang G, Fu J and Wu Q: Matrix metalloproteinase-14 (MMP-14) downregulation inhibits esophageal squamous cell carcinoma cell migration, invasion, and proliferation. Thorac Cancer. 11:3168–3174. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li M, Li S, Zhou L, Yang L, Wu X, Tang B, Xie S, Fang L, Zheng S and Hong T: Immune Infiltration of MMP14 in pan cancer and its prognostic effect on tumors. Front Oncol. 11:7176062021. View Article : Google Scholar : PubMed/NCBI | |
Fan QC, Tian H, Wang Y and Liu XB: Integrin-α5 promoted the progression of oral squamous cell carcinoma and modulated PI3K/AKT signaling pathway. Arch Oral Biol. 101:85–91. 2019. View Article : Google Scholar : PubMed/NCBI | |
Park SJ, Min HJ, Yoon C, Kim SH, Kim JH and Lee SY: Integrin β1 regulates the perineural invasion and radioresistance of oral squamous carcinoma cells by modulating cancer cell stemness. Cell Signal. 110:1108082023. View Article : Google Scholar : PubMed/NCBI | |
Ramos DM, But M, Regezi J, Schmidt BL, Atakilit A, Dang D, Ellis D, Jordan R and Li X: Expression of integrin beta 6 enhances invasive behavior in oral squamous cell carcinoma. Matrix Biol. 21:297–307. 2002. View Article : Google Scholar : PubMed/NCBI | |
Ishida Y, Shintani T, Nobumoto T, Sakurai S, Hamana T, Yanamoto S and Hayashido Y: Interaction of Integrin αvβ8 With Type I collagen promotes squamous cell carcinoma cell motility via RAC1 activation. Anticancer Res. 43:4833–4841. 2023. View Article : Google Scholar : PubMed/NCBI | |
Thomas GJ, Jones J and Speight PM: Integrins and oral cancer. Oral Oncol. 33:381–388. 1997. View Article : Google Scholar : PubMed/NCBI | |
Hou S, Hao X, Li J, Weng S, Wang J, Zhao T, Li W, Hu X, Deng B, Gu J and Hang Q: TM4SF1 promotes esophageal squamous cell carcinoma metastasis by interacting with integrin α6. Cell Death Dis. 13:6092022. View Article : Google Scholar : PubMed/NCBI | |
Xie YH, Ran LQ, Wu ZY, Sun C, Xu XE, Zou HY, Fang WK and Xie JJ: Role of Integrin β1 in the progression and chemo-resistance of esophageal squamous cell carcinoma. J Cancer. 13:2074–2085. 2022. View Article : Google Scholar : PubMed/NCBI | |
Xie JJ, Guo JC, Wu ZY, Xu XE, Wu JY, Chen B, Ran LQ, Liao LD, Li EM and Xu LY: Integrin α5 promotes tumor progression and is an independent unfavorable prognostic factor in esophageal squamous cell carcinoma. Hum Pathol. 48:69–75. 2016. View Article : Google Scholar : PubMed/NCBI | |
Dmello C, Sawant S, Alam H, Gangadaran P, Tiwari R, Dongre H, Rana N, Barve S, Costea DE, Chaukar D, et al: Vimentin-mediated regulation of cell motility through modulation of beta4 integrin protein levels in oral tumor derived cells. Int J Biochem Cell Biol. 70:161–172. 2016. View Article : Google Scholar : PubMed/NCBI | |
Liu S, Liao G and Li G: Regulatory effects of COL1A1 on apoptosis induced by radiation in cervical cancer cells. Cancer Cell Int. 17:732017. View Article : Google Scholar : PubMed/NCBI | |
Geng Q, Shen Z, Li L and Zhao J: COL1A1 is a prognostic biomarker and correlated with immune infiltrates in lung cancer. PeerJ. 9:e111452021. View Article : Google Scholar : PubMed/NCBI | |
Lin P, Tian P, Pang J, Lai L, He G, Song Y and Zheng Y: Clinical significance of COL1A1 and COL1A2 expression levels in hypopharyngeal squamous cell carcinoma. Oncol Lett. 20:803–809. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li G, Jiang W, Kang Y, Yu X, Zhang C and Feng Y: High expression of collagen 1A2 promotes the proliferation and metastasis of esophageal cancer cells. Ann Transl Med. 8:16722020. View Article : Google Scholar : PubMed/NCBI | |
Shen Y, Li X, Wang D, Zhang L, Li X, Su L, Fan X and Yang X: COL3A1: Potential prognostic predictor for head and neck cancer based on immune-microenvironment alternative splicing. Cancer Med. 12:4882–4894. 2023. View Article : Google Scholar : PubMed/NCBI | |
Tian X, Sun J, Li C and Zhang K: COL4A1 promotes the proliferation and migration of oral squamous cell carcinoma cells by binding to NID1. Exp Ther Med. 25:1762023. View Article : Google Scholar : PubMed/NCBI | |
Di YB, Bao Y, Guo J, Liu W, Zhang SX, Zhang GH and Li TK: COL11A1 as a potential prognostic target for oral squamous cell carcinoma. Medicine (Baltimore). 101:e309892022. View Article : Google Scholar : PubMed/NCBI | |
Sok JC, Lee JA, Dasari S, Joyce S, Contrucci SC, Egloff AM, Trevelline BK, Joshi R, Kumari N, Grandis JR and Thomas SM: Collagen type XI α1 facilitates head and neck squamous cell cancer growth and invasion. Br J Cancer. 109:3049–3056. 2013. View Article : Google Scholar : PubMed/NCBI | |
Galiger C, Löffek S, Stemmler MP, Kroeger JK, Mittapalli VR, Fauth L, Esser PR, Kern JS, Meiss F, Laßmann S, et al: Targeting of cell surface proteolysis of collagen XVII impedes squamous cell carcinoma progression. Mol Ther. 26:17–30. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Jung SN, Oh C, Lee K, Won HR, Chang JW, Kim JM and Koo BS: LAMB3 is associated with disease progression and cisplatin cytotoxic sensitivity in head and neck squamous cell carcinoma. Eur J Surg Oncol. 45:359–365. 2019. View Article : Google Scholar : PubMed/NCBI | |
Marangon Junior H, Rocha VN, Leite CF, de Aguiar MC, Souza PE and Horta MC: Laminin-5 gamma 2 chain expression is associated with intensity of tumor budding and density of stromal myofibroblasts in oral squamous cell carcinoma. J Oral Pathol Med. 43:199–204. 2014. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Zhou J, Lu J, Xiong H, Shi X and Gong L: Significance of CD44 expression in head and neck cancer: A systemic review and meta-analysis. BMC Cancer. 14:152014. View Article : Google Scholar : PubMed/NCBI | |
Mirhashemi M, Sadeghi M, Ghazi N, Saghravanian N, Dehghani M and Aminian A: Prognostic value of CD44 expression in oral squamous cell carcinoma: A meta-analysis. Ann Diagn Pathol. 67:1522132023. View Article : Google Scholar : PubMed/NCBI | |
Sawant S, Ahire C, Dongre H, Joshi S, Jamghare S, Rane P, Kane S and Chaukar D: Prognostic significance of elevated serum CD44 levels in patients with oral squamous cell carcinoma. J Oral Pathol Med. 47:665–673. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kudo Y, Ogawa I, Kitajima S, Kitagawa M, Kawai H, Gaffney PM, Miyauchi M and Takata T: Periostin promotes invasion and anchorage-independent growth in the metastatic process of head and neck cancer. Cancer Res. 66:6928–6935. 2006. View Article : Google Scholar : PubMed/NCBI | |
Maleš J, Mihalj H, Šestak A, Kralik K and Smolić M: Osteopontin levels in patients with squamous metastatic head and neck cancer. Medicina (Kaunas). 57:1852021. View Article : Google Scholar : PubMed/NCBI | |
Li J, Wang X, Zheng K, Liu Y, Li J and Wang S, Liu K, Song X, Li N, Xie S and Wang S: The clinical significance of collagen family gene expression in esophageal squamous cell carcinoma. PeerJ. 7:e77052019. View Article : Google Scholar : PubMed/NCBI | |
Zhou J, Yang Y, Zhang H, Luan S, Xiao X, Li X, Fang P, Shang Q, Chen L, Zeng X and Yuan Y: Overexpressed COL3A1 has prognostic value in human esophageal squamous cell carcinoma and promotes the aggressiveness of esophageal squamous cell carcinoma by activating the NF-κB pathway. Biochem Biophys Res Commun. 613:193–200. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhang B, Zhang C, Yang X, Chen Y, Zhang H, Liu J and Wu Q: Cytoplasmic collagen XIαI as a prognostic biomarker in esophageal squamous cell carcinoma. Cancer Biol Ther. 19:364–372. 2018. View Article : Google Scholar : PubMed/NCBI | |
Meng X, Chen X, Lu P, Ma W, Yue D, Song L and Fan Q: MicroRNA-202 inhibits tumor progression by targeting LAMA1 in esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 473:821–827. 2016. View Article : Google Scholar : PubMed/NCBI | |
Shen XM, Wu YP, Feng YB, Luo ML, Du XL, Zhang Y, Cai Y, Xu X, Han YL, Zhang X, et al: Interaction of MT1-MMP and laminin-5gamma2 chain correlates with metastasis and invasiveness in human esophageal squamous cell carcinoma. Clin Exp Metastasis. 24:541–550. 2007. View Article : Google Scholar : PubMed/NCBI | |
Liang Y, Chen X, Wu Y, Li J, Zhang S, Wang K, Guan X, Yang K and Bai Y: LncRNA CASC9 promotes esophageal squamous cell carcinoma metastasis through upregulating LAMC2 expression by interacting with the CREB-binding protein. Cell Death Differ. 25:1980–1995. 2018. View Article : Google Scholar : PubMed/NCBI | |
Fang L, Che Y, Zhang C, Huang J, Lei Y, Lu Z, Sun N and He J: LAMC1 upregulation via TGFβ induces inflammatory cancer-associated fibroblasts in esophageal squamous cell carcinoma via NF-κB-CXCL1-STAT3. Mol Oncol. 15:3125–3146. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kamil Mohammed Al-Mosawi A, Cheshomi H, Hosseinzadeh A and M Matin M: Prognostic and Clinical Value of CD44 and CD133 in Esophageal Cancer: A Systematic Review and Meta-analysis. Iran J Allergy Asthma Immunol. 19:105–116. 2020.PubMed/NCBI | |
Miyako S, Koma YI, Nakanishi T, Tsukamoto S, Yamanaka K, Ishihara N, Azumi Y, Urakami S, Shimizu M, Kodama T, et al: Periostin in cancer-associated fibroblasts promotes esophageal squamous cell carcinoma progression by enhancing cancer and stromal cell migration. Am J Pathol. 194:828–848. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen B, Liang S, Guo H, Xu L, Li J and Peng J: OPN promotes cell proliferation and invasion through NF-κB in human esophageal squamous cell carcinoma. Genet Res (Camb). 2022:31548272022. View Article : Google Scholar : PubMed/NCBI | |
Chiu TJ, Lu HI, Chen CH, Huang WT, Wang YM, Lin WC and Li SH: Osteopontin expression is associated with the poor prognosis in patients with locally advanced esophageal squamous cell carcinoma receiving preoperative chemoradiotherapy. Biomed Res Int. 2018:90982152018. View Article : Google Scholar : PubMed/NCBI | |
Duan Y, Liu G, Sun Y, Wu J, Xiong Z, Jin T and Chen M: Collagen type VI α5 gene variations may predict the risk of lung cancer development in Chinese Han population. Sci Rep. 10:50102020. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Sun Y, Guo Z and Liu H: COL3A1 overexpression associates with poor prognosis and cisplatin resistance in lung cancer. Balkan Med J. 39:393–400. 2022. View Article : Google Scholar : PubMed/NCBI | |
Voiles L, Lewis DE, Han L, Lupov IP, Lin TL, Robertson MJ, Petrache I and Chang HC: Overexpression of type VI collagen in neoplastic lung tissues. Oncol Rep. 32:1897–1904. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lee CS, Siprashvili Z, Mah A, Bencomo T, Elcavage LE, Che Y, Shenoy RM, Aasi SZ and Khavari PA: Mutant collagen COL11A1 enhances cancerous invasion. Oncogene. 40:6299–6307. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu M, Cai R, Wang T, Yang X, Wang M, Kuang Z, Xie Y, Zhang J and Zheng Y: LAMC2 promotes the proliferation of cancer cells and induce infiltration of macrophages in non-small cell lung cancer. Ann Transl Med. 9:13922021. View Article : Google Scholar : PubMed/NCBI | |
Akashi T, Ito E, Eishi Y, Koike M, Nakamura K and Burgeson RE: Reduced Expression of Laminin alpha3 and alpha5 Chains in Non-small Cell Lung Cancers. Jpn J Cancer Res. 92:293–301. 2001. View Article : Google Scholar : PubMed/NCBI | |
Rousselle P and Scoazec JY: Laminin 332 in cancer: When the extracellular matrix turns signals from cell anchorage to cell movement. Semin Cancer Biol. 62:149–165. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jiang H, Zhao W and Shao W: Prognostic value of CD44 and CD44v6 expression in patients with non-small cell lung cancer: meta-analysis. Tumour Biol. 35:7383–7389. 2014. View Article : Google Scholar : PubMed/NCBI | |
Xu CH, Wang W, Lin Y, Qian LH, Zhang XW, Wang QB and Yu LK: Diagnostic and prognostic value of serum periostin in patients with non-small cell lung cancer. Oncotarget. 8:18746–18753. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wang W, Wang S and Zhang M: Evaluation of kininogen 1, osteopontin and α-1-antitrypsin in plasma, bronchoalveolar lavage fluid and urine for lung squamous cell carcinoma diagnosis. Oncol Lett. 19:2785–2792. 2020.PubMed/NCBI | |
Hou T, Tong C, Kazobinka G, Zhang W, Huang X, Huang Y and Zhang Y: Expression of COL6A1 predicts prognosis in cervical cancer patients. Am J Transl Res. 8:2838–2844. 2016.PubMed/NCBI | |
Skyldberg B, Salo S, Eriksson E, Aspenblad U, Moberger B, Tryggvason K and Auer G: Laminin-5 as a Marker of Invasiveness in Cervical Lesions. J Natl Cancer Inst. 91:1882–1887. 1999. View Article : Google Scholar : PubMed/NCBI | |
Noel JC, Fernandez-Aguilar S, Fayt I, Buxant F, Ansion MH, Simon P and Anaf V: Laminin-5γ2 chain expression in cervical intraepithelial neoplasia and invasive cervical carcinoma. Acta Obstet Gynecol Scand. 84:1119–1123. 2005. View Article : Google Scholar : PubMed/NCBI | |
Kainz C, Kohlberger P, Tempfer C, Sliutz G, Gitsch G, Reinthaller A and Breitenecker G: Prognostic value of CD44 splice variants in human stage III cervical cancer. Eur J Cancer. 31:1706–1709. 1995. View Article : Google Scholar : PubMed/NCBI | |
Wei WF, Chen XJ, Liang LJ, Yu L, Wu XG, Zhou CF, Wang ZC, Fan LS, Hu Z, Liang L and Wang W: Periostin+cancer-associated fibroblasts promote lymph node metastasis by impairing the lymphatic endothelial barriers in cervical squamous cell carcinoma. Mol Oncol. 15:210–227. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gillot L, Lebeau A, Baudin L, Pottier C, Louis T, Durré T, Longuespée R, Mazzucchelli G, Nizet C, Blacher S, et al: Periostin in lymph node pre-metastatic niches governs lymphatic endothelial cell functions and metastatic colonization. Cell Mol Life Sci. 79:2952022. View Article : Google Scholar : PubMed/NCBI | |
Qin S, Yi L, Liang Y, Chen Y, Wang W, Liao Y, Zhang C, Huang H, Huang J and Yao S: Biological and Clinicopathological Characteristics of OPN in Cervical Cancers. Front Genet. 13:8365092022. View Article : Google Scholar : PubMed/NCBI | |
Castor MDGFC, Torres LC, Mello RJV, Natal RA and Vassallo J: Study on collagen parameters in vulvar cancer and preneoplastic lesions by Second Harmonic Generation microscopy. Sci Rep. 10:55682020. View Article : Google Scholar : PubMed/NCBI | |
Holthoff ER, Byrum SD, Mackintosh SG, Kelly T, Tackett AJ, Quick CM and Post SR: Vulvar squamous cell carcinoma aggressiveness is associated with differential expression of collagen and STAT1. Clin Proteomics. 14:402017. View Article : Google Scholar : PubMed/NCBI | |
Wu Z, Shen Y, Gong K, Wu Z, Zhang T, Zhang X and Li S: Increased osteopontin expression is associated with progression from vulvar precancerous lesions to vulvar squamous cell carcinoma. Arch Gynecol Obstet. 289:637–644. 2014. View Article : Google Scholar : PubMed/NCBI | |
Sgambato A, Tarquini E, Resci F, De Paola B, Faraglia B, Camerini A, Rettino A, Migaldi M, Cittadini A and Zannoni GF: Aberrant expression of alpha-dystroglycan in cervical and vulvar cancer. Gynecol Oncol. 103:397–404. 2006. View Article : Google Scholar : PubMed/NCBI | |
Brockbank EC, Bridges J, Marshall CJ and Sahai E: Integrin beta1 is required for the invasive behaviour but not proliferation of squamous cell carcinoma cells in vivo. Br J Cancer. 92:102–112. 2005. View Article : Google Scholar : PubMed/NCBI | |
Hefler LA, Concin N, Mincham D, Thompson J, Swarte NB, van Eijkeren MA, Sie-Go DM, Hammond I, McCartney AJ, Tempfer CB and Speiser P: The prognostic value of immunohistochemically detected CD44v3 and CD44v6 expression in patients with surgically staged vulvar carcinoma: A multicenter study. Cancer. 94:125–130. 2002. View Article : Google Scholar : PubMed/NCBI | |
Johansson N, Vaalamo M, Grénman S, Hietanen S, Klemi P, Saarialho-Kere U and Kähäri VM: Collagenase-3 (MMP-13) is expressed by tumor cells in invasive vulvar squamous cell carcinomas. Am J Pathol. 154:469–480. 1999. View Article : Google Scholar : PubMed/NCBI | |
Bovo AC, da Silva ID, Takita LC, Fochi J, Stávale JN, Marks G and de Lima GR: A comparative study of MMP-2 in vulvar neoplasms. Gynecol Oncol. 93:454–457. 2004. View Article : Google Scholar : PubMed/NCBI | |
Hua H, Li M, Luo T, Yin Y and Jiang Y: Matrix metalloproteinases in tumorigenesis: An evolving paradigm. Cell Mol Life Sci. 68:3853–3868. 2011. View Article : Google Scholar : PubMed/NCBI | |
Kerkelä E, Ala-aho R, Klemi P, Grénman S, Shapiro SD, Kähäri VM and Saarialho-Kere U: Metalloelastase (MMP-12) expression by tumour cells in squamous cell carcinoma of the vulva correlates with invasiveness, while that by macrophages predicts better outcome. J Pathol. 198:258–269. 2002. View Article : Google Scholar : PubMed/NCBI | |
Goepel C, Stoerer S and Koelbl H: Tenascin in preinvasive lesions of the vulva and vulvar cancer. Anticancer Res. 23:4587–4591. 2003.PubMed/NCBI | |
Surico N, Priori L, Savoia P, Cremona O and Marchisio PC: Distribution of integrins and extracellular matrix proteins in vulvar squamous cell carcinomas. Eur J Gynaecol Oncol. 16:147–154. 1995.PubMed/NCBI | |
Hellman K, Hellström AC, Silfverswärd C, Salo S, Aspenblad U, Nilsson B, Frankendal B, Tryggvasson K and Auer G: Cancer of the vagina: Laminin-5gamma2 chain expression and prognosis. Int J Gynecol Cancer. 10:391–396. 2000. View Article : Google Scholar : PubMed/NCBI | |
Theocharis AD, Manou D and Karamanos NK: The extracellular matrix as a multitasking player in disease. FEBS J. 286:2830–2869. 2019. View Article : Google Scholar : PubMed/NCBI | |
Diao B and Yang P: Comprehensive analysis of the expression and prognosis for laminin genes in ovarian cancer. Pathol Oncol Res. 27:16098552021. View Article : Google Scholar : PubMed/NCBI | |
Jing J, Lien CF, Sharma S, Rice J, Brennan PA and Górecki DC: Aberrant expression, processing and degradation of dystroglycan in squamous cell carcinomas. Eur J Cancer. 40:2143–2151. 2004. View Article : Google Scholar : PubMed/NCBI | |
Sgambato A and Brancaccio A: The dystroglycan complex: From biology to cancer. J Cell Physiol. 205:163–169. 2005. View Article : Google Scholar : PubMed/NCBI | |
Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuña JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N and Martinez-Fierro ML: the roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci. 21:97392020. View Article : Google Scholar : PubMed/NCBI | |
Lu P, Weaver VM and Werb Z: The extracellular matrix: A dynamic niche in cancer progression. J Cell Biol. 196:395–406. 2012. View Article : Google Scholar : PubMed/NCBI | |
Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, et al: A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 20:174–186. 2020. View Article : Google Scholar : PubMed/NCBI | |
Costea DE, Hills A, Osman AH, Thurlow J, Kalna G, Huang X, Pena Murillo C, Parajuli H, Suliman S, Kulasekara KK, et al: Identification of two distinct carcinoma-associated fibroblast subtypes with differential tumor-promoting abilities in oral squamous cell carcinoma. Cancer Res. 73:3888–3901. 2013. View Article : Google Scholar : PubMed/NCBI | |
Obradovic A, Graves D, Korrer M, Wang Y, Roy S, Naveed A, Xu Y, Luginbuhl A, Curry J, Gibson M, et al: Immunostimulatory cancer-associated fibroblast subpopulations can predict immunotherapy response in head and neck cancer. Clin Cancer Res. 28:2094–2109. 2022. View Article : Google Scholar : PubMed/NCBI | |
Puram SV, Tirosh I, Parikh AS, Yizhak K, Gillespie S, Rodman C, Luo CL, Mroz EA, Emerick KS, Deschler DG, et al: Single-Cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell. 171:1611–1624.e24. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hu C, Zhang Y, Wu C and Huang Q: Heterogeneity of cancer-associated fibroblasts in head and neck squamous cell carcinoma: Opportunities and challenges. Cell Death Discov. 9:1242023. View Article : Google Scholar : PubMed/NCBI | |
Mirkeshavarz M, Ganjibakhsh M, Aminishakib P, Farzaneh P, Mahdavi N, Vakhshiteh F, Karimi A, Gohari NS, Kamali F, Kharazifard MJ, et al: Interleukin-6 secreted by oral cancer-associated fibroblast accelerated VEGF expression in tumor and stroma cells. Cell Mol Biol (Noisy-le-grand). 63:131–136. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bae JY, Kim EK, Yang DH, Zhang X, Park YJ, Lee DY, Che CM and Kim J: Reciprocal interaction between carcinoma-associated fibroblasts and squamous carcinoma cells through interleukin-1α induces cancer progression. Neoplasia. 16:928–938. 2014. View Article : Google Scholar : PubMed/NCBI | |
Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, Corbo V, Oni TE, Hearn SA, Lee EJ, et al: Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 214:579–596. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lavie D, Ben-Shmuel A, Erez N and Scherz-Shouval R: Cancer-associated fibroblasts in the single-cell era. Nat Cancer. 3:793–807. 2022. View Article : Google Scholar : PubMed/NCBI | |
El Herch I, Tornaas S, Dongre HN and Costea DE: Heterogeneity of cancer-associated fibroblasts and tumor-promoting roles in head and neck squamous cell carcinoma. Front Mol Biosci. 11:13400242024. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Zhang L, Zhu Y, Zhao D, Zhang J, Zhu Y, Pang J, Xiao Y, Wu Q, Wang Y and Zhan Q: AKT2(S128)/CCTα(S315/319/323)-positive cancer-associated fibroblasts (CAFs) mediate focal adhesion kinase (FAK) inhibitors resistance via secreting phosphatidylcholines (PCs). Signal Transduct Target Ther. 9:212024. View Article : Google Scholar : PubMed/NCBI | |
Li W, Xu T, Jin H, Li M and Jia Q: Emerging role of cancer-associated fibroblasts in esophageal squamous cell carcinoma. Pathol Res Pract. 253:1550022024. View Article : Google Scholar : PubMed/NCBI | |
Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, Xu Q, Shi J, Lu E, Chen W and Zhang J: Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 20:122019. View Article : Google Scholar : PubMed/NCBI | |
Huang W, Zhang L, Yang M, Wu X, Wang X, Huang W, Yuan L, Pan H, Wang Y, Wang Z, et al: Cancer-associated fibroblasts promote the survival of irradiated nasopharyngeal carcinoma cells via the NF-κB pathway. J Exp Clin Cancer Res. 40:872021. View Article : Google Scholar : PubMed/NCBI | |
Li X, González-Maroto C and Tavassoli M: Crosstalk between CAFs and tumour cells in head and neck cancer. Cell Death Discov. 10:3032024. View Article : Google Scholar : PubMed/NCBI | |
Dongre H, Rana N, Fromreide S, Rajthala S, Bøe Engelsen I, Paradis J, Gutkind JS, Vintermyr OK, Johannessen AC, Bjørge L and Costea DE: Establishment of a novel cancer cell line derived from vulvar carcinoma associated with lichen sclerosus exhibiting a fibroblast-dependent tumorigenic potential. Exp Cell Res. 386:1116842020. View Article : Google Scholar : PubMed/NCBI | |
Milacic M, Beavers D, Conley P, Gong C, Gillespie M, Griss J, Haw R, Jassal B, Matthews L, May B, et al: The reactome pathway knowledgebase 2024. Nucleic Acids Res. 52((D1)): D672–D678. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, Li B and Liu XS: TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 48:W509–W514. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cancer Genome Atlas Network, . Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517:576–582. 2015. View Article : Google Scholar : PubMed/NCBI | |
Cancer Genome Atlas Research Network and Albert Einstein College of Medicine; Analytical Biological Services; Barretos Cancer Hospital; Baylor College of Medicine; Beckman Research Institute of City of Hope; Buck Institute for Research on Aging; Canada's Michael Smith Genome Sciences Centre; Harvard Medical School; Helen F. Graham Cancer Center &Research Institute at Christiana Care Health Services et al., . Integrated genomic and molecular characterization of cervical cancer. Nature. 543:378–384. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cancer Genome Atlas Research Network; Analysis Working Group; Asan University; BC Cancer Agency; Brigham and Women's Hospital; Broad Institute; Brown University; Case Western Reserve University; Dana-Farber Cancer Institute; Duke University et al., . Integrated genomic characterization of oesophageal carcinoma. Nature. 541:169–175. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cancer Genome Atlas Research Network, . Comprehensive genomic characterization of squamous cell lung cancers. Nature. 489:519–525. 2012. View Article : Google Scholar : PubMed/NCBI | |
Bergonzini C, Kroese K, Zweemer AJM and Danen EHJ: Targeting integrins for cancer therapy-disappointments and opportunities. Front Cell Dev Biol. 10:8638502022. View Article : Google Scholar : PubMed/NCBI | |
Winer A, Adams S and Mignatti P: Matrix metalloproteinase inhibitors in cancer therapy: Turning past failures into future successes. Mol Cancer Ther. 17:1147–1155. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yan Z, Hu X, Tang B and Deng F: Role of osteopontin in cancer development and treatment. Heliyon. 9:e210552023. View Article : Google Scholar : PubMed/NCBI | |
Zagani R, Hamzaoui N, Cacheux W, de Reyniès A, Terris B, Chaussade S, Romagnolo B, Perret C and Lamarque D: Cyclooxygenase-2 inhibitors down-regulate osteopontin and Nr4A2-new therapeutic targets for colorectal cancers. Gastroenterology. 37:1358–1366.e1-e3. 2009. View Article : Google Scholar | |
Sroka TC, Pennington ME and Cress AE: Synthetic D-amino acid peptide inhibits tumor cell motility on laminin-5. Carcinogenesis. 27:1748–1757. 2006. View Article : Google Scholar : PubMed/NCBI | |
Tran M, Rousselle P, Nokelainen P, Tallapragada S, Nguyen NT, Fincher EF and Marinkovich MP: Targeting a tumor-specific laminin domain critical for human carcinogenesis. Cancer Res. 68:2885–2894. 2008. View Article : Google Scholar : PubMed/NCBI | |
Heider KH, Sproll M, Susani S, Patzelt E, Beaumier P, Ostermann E, Ahorn H and Adolf GR: Characterization of a high-affinity monoclonal antibody specific for CD44v6 as candidate for immunotherapy of squamous cell carcinomas. Cancer Immunol Immunother. 43:245–253. 1996. View Article : Google Scholar : PubMed/NCBI | |
Sandström K, Nestor M, Ekberg T, Engström M, Anniko M and Lundqvist H: Targeting CD44v6 expressed in head and neck squamous cell carcinoma: Preclinical characterization of an 111In-labeled monoclonal antibody. Tumour Biol. 29:137–144. 2008. View Article : Google Scholar : PubMed/NCBI | |
Song K, Yu Z, Zu X, Li G, Hu Z and Xue Y: Collagen remodeling along cancer progression providing a novel opportunity for cancer diagnosis and treatment. Int J Mol Sci. 23:105092022. View Article : Google Scholar : PubMed/NCBI | |
Singh B, Sims H, Trueheart I, Simpson K, Wang KC, Patzkowsky K, Wegman T, Soma JM, Dixon R, Jayes F, et al: A Phase I clinical trial to assess safety and tolerability of injectable collagenase in women with symptomatic uterine fibroids. Reprod Sci. 28:2699–2709. 2021. View Article : Google Scholar : PubMed/NCBI | |
Oo KK, Kamolhan T, Soni A, Thongchot S, Mitrpant C, O-Charoenrat P, Thuwajit C and Thuwajit P: Development of an engineered peptide antagonist against periostin to overcome doxorubicin resistance in breast cancer. BMC Cancer. 21:652021. View Article : Google Scholar : PubMed/NCBI | |
Nakazawa Y, Taniyama Y, Sanada F, Morishita R, Nakamori S, Morimoto K, Yeung KT and Yang J: Periostin blockade overcomes chemoresistance via restricting the expansion of mesenchymal tumor subpopulations in breast cancer. Sci Rep. 8:40132018. View Article : Google Scholar : PubMed/NCBI | |
Zhu M, Saxton RE, Ramos L, Chang DD, Karlan BY, Gasson JC and Slamon DJ: Neutralizing monoclonal antibody to periostin inhibits ovarian tumor growth and metastasis. Mol Cancer Ther. 10:1500–1508. 2011. View Article : Google Scholar : PubMed/NCBI | |
Lee YJ, Kim IS, Park SA, Kim Y, Lee JE, Noh DY, Kim KT, Ryu SH and Suh PG: Periostin-binding DNA aptamer inhibits breast cancer growth and metastasis. Mol Ther. 21:1004–1013. 2013. View Article : Google Scholar : PubMed/NCBI | |
Liu GX, Xi HQ, Sun XY and Wei B: Role of periostin and its antagonist PNDA-3 in gastric cancer metastasis. World J Gastroenterol. 21:2605–2613. 2015. View Article : Google Scholar : PubMed/NCBI |