The Tumor Microenvironment: A Dynamic Ecosystem Sculpting Cancer Evolution and Therapeutic Response

Abstract

The tumor microenvironment (TME) has emerged as a critical determinant of cancer progression, metastasis, and therapeutic resistance. This complex ecosystem, comprising cancer cells, stromal cells, immune cells, vasculature, and extracellular matrix (ECM), dynamically interacts to shape tumor behavior. Beyond simply providing a supportive niche, the TME actively contributes to genetic instability, epigenetic modifications, and metabolic reprogramming within cancer cells, accelerating their adaptation and survival. This review delves into the multifaceted roles of key TME components, highlighting their intricate signaling pathways and their influence on cancer cell plasticity. We explore the emerging concept of the TME as a driver of intratumoral heterogeneity and its implications for targeted therapies. Furthermore, we discuss novel strategies that disrupt pro-tumorigenic TME interactions, including approaches that remodel the ECM, modulate immune cell function, and normalize tumor vasculature. Finally, we address the challenges of translating TME-targeted therapies into clinical success, emphasizing the need for personalized approaches that account for the inherent complexity and variability of the TME across different cancer types and stages.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

1. Introduction

The classical view of cancer as a cell-autonomous disease has undergone a paradigm shift, with increasing recognition of the crucial role of the tumor microenvironment (TME). The TME is not merely a passive bystander; it is a dynamic and interactive ecosystem that actively participates in all stages of cancer development, from initiation and growth to invasion, metastasis, and therapeutic resistance. This complex milieu comprises diverse cellular and non-cellular components, including cancer cells, stromal cells (fibroblasts, endothelial cells, pericytes), immune cells (both innate and adaptive), vasculature, and the extracellular matrix (ECM). These components interact through intricate signaling networks, mediated by soluble factors (cytokines, chemokines, growth factors), cell-cell contacts, and ECM remodeling. The TME provides essential nutrients, growth factors, and structural support to cancer cells, while simultaneously shaping their genetic and epigenetic landscape. In essence, the TME is a selective force that drives cancer evolution and determines therapeutic response.

The importance of the TME is underscored by several key observations. First, many cancers exhibit significant stromal infiltration, indicating an active recruitment and reprogramming of surrounding tissues. Second, the TME can promote angiogenesis, providing cancer cells with access to oxygen and nutrients, thereby facilitating tumor growth and metastasis. Third, the TME can suppress anti-tumor immune responses, allowing cancer cells to evade immune surveillance and destruction. Fourth, the TME can contribute to drug resistance by sequestering drugs, inducing epithelial-mesenchymal transition (EMT), and activating survival pathways in cancer cells. Finally, targeting the TME has shown promising results in preclinical and clinical studies, suggesting that it represents a valuable therapeutic avenue.

However, the TME is not a monolithic entity. Its composition and architecture vary significantly across different cancer types, stages, and even within different regions of the same tumor. This heterogeneity poses a major challenge for developing effective TME-targeted therapies. A deeper understanding of the specific TME components and their interactions in different cancer contexts is crucial for designing personalized strategies that disrupt pro-tumorigenic signaling and restore anti-tumor immunity.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2. Key Components of the Tumor Microenvironment

2.1 Cancer-Associated Fibroblasts (CAFs)

Cancer-associated fibroblasts (CAFs) are one of the most abundant stromal cell types in many solid tumors. These activated fibroblasts are characterized by their increased expression of α-smooth muscle actin (α-SMA), fibroblast activation protein (FAP), and other markers. CAFs play a multifaceted role in cancer progression, influencing tumor growth, angiogenesis, ECM remodeling, and immune modulation.

  • ECM Remodeling: CAFs are a major source of ECM components, including collagen, fibronectin, and laminin. They also secrete matrix metalloproteinases (MMPs) and other ECM-degrading enzymes, which remodel the ECM and facilitate cancer cell invasion and metastasis. However, the role of ECM remodeling is not always straightforward. While some ECM modifications promote tumor progression, others can restrict cancer cell migration and inhibit angiogenesis. This duality highlights the complexity of CAF-ECM interactions.

  • Growth Factor Production: CAFs secrete a wide range of growth factors, such as hepatocyte growth factor (HGF), epidermal growth factor (EGF), and platelet-derived growth factor (PDGF), which stimulate cancer cell proliferation, survival, and migration. These growth factors can also activate downstream signaling pathways, such as the PI3K/AKT and MAPK pathways, which further enhance cancer cell growth and survival.

  • Angiogenesis: CAFs promote angiogenesis by secreting vascular endothelial growth factor (VEGF) and other pro-angiogenic factors. They can also directly interact with endothelial cells to stimulate their proliferation and migration, leading to the formation of new blood vessels. The newly formed vessels are often abnormal and leaky, contributing to tumor hypoxia and further promoting tumor progression.

  • Immune Modulation: CAFs can suppress anti-tumor immune responses by secreting immunosuppressive cytokines, such as transforming growth factor-β (TGF-β) and interleukin-10 (IL-10). They can also recruit immunosuppressive immune cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), to the TME. These immune cells further inhibit anti-tumor immunity and promote tumor growth.

The origin of CAFs is diverse and includes resident fibroblasts, bone marrow-derived cells, and epithelial cells undergoing epithelial-mesenchymal transition (EMT). The specific origin of CAFs can influence their phenotype and function, adding another layer of complexity to their role in cancer progression. Targeting CAFs has emerged as a promising therapeutic strategy, but the heterogeneous nature of CAFs and their complex interactions with cancer cells present significant challenges.

2.2 Immune Cells

The immune system plays a dual role in cancer, acting as both a tumor suppressor and a tumor promoter. On one hand, immune cells, such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, can recognize and eliminate cancer cells. On the other hand, immune cells, such as Tregs and MDSCs, can suppress anti-tumor immunity and promote tumor growth.

  • Anti-tumor Immunity: CTLs are the primary mediators of anti-tumor immunity. They recognize cancer cells that express tumor-associated antigens (TAAs) and kill them through the release of cytotoxic granules or by activating death receptor pathways. NK cells can also kill cancer cells, particularly those that have lost expression of MHC class I molecules. The balance between CTL activation and suppression is critical for determining the outcome of anti-tumor immunity.

  • Immune Evasion: Cancer cells can evade immune surveillance through various mechanisms, including downregulation of MHC class I expression, secretion of immunosuppressive cytokines, and recruitment of immunosuppressive immune cells. The TME plays a key role in promoting immune evasion. For example, CAFs can secrete TGF-β, which inhibits CTL activation and promotes Treg differentiation. Tumor-associated macrophages (TAMs) can also suppress anti-tumor immunity by secreting IL-10 and arginase.

  • Tumor-Promoting Inflammation: Chronic inflammation can promote cancer development by providing a permissive environment for tumor growth and metastasis. Inflammatory cells, such as macrophages and neutrophils, can release reactive oxygen species (ROS) and other factors that damage DNA and promote genomic instability. They can also secrete growth factors and cytokines that stimulate cancer cell proliferation and angiogenesis.

  • Immune Checkpoint Blockade: Immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 antibodies, have revolutionized cancer therapy by blocking inhibitory signaling pathways that suppress anti-tumor immunity. These therapies have shown remarkable efficacy in certain cancers, but they are not effective in all patients. Understanding the mechanisms of resistance to immune checkpoint blockade is a major focus of current research. The TME plays a crucial role in determining the response to immune checkpoint inhibitors. For example, tumors with high levels of T cell infiltration and low levels of immunosuppressive cells are more likely to respond to these therapies.

The composition and function of immune cells within the TME are highly variable and depend on the specific cancer type, stage, and genetic background of the patient. Targeting the TME to enhance anti-tumor immunity and overcome immune evasion represents a promising therapeutic strategy.

2.3 Vasculature

The tumor vasculature provides cancer cells with essential oxygen and nutrients and serves as a conduit for metastasis. However, tumor blood vessels are often abnormal and dysfunctional, characterized by their irregular structure, increased permeability, and impaired perfusion. This abnormal vasculature contributes to tumor hypoxia, which promotes tumor growth, angiogenesis, and metastasis.

  • Angiogenesis: Angiogenesis, the formation of new blood vessels, is essential for tumor growth and metastasis. Cancer cells secrete VEGF and other pro-angiogenic factors that stimulate endothelial cell proliferation, migration, and tube formation. The newly formed vessels are often disorganized and leaky, leading to increased interstitial pressure and impaired drug delivery.

  • Vascular Normalization: Vascular normalization is a therapeutic strategy aimed at improving the structure and function of tumor blood vessels. This can be achieved by targeting VEGF signaling with anti-VEGF antibodies or tyrosine kinase inhibitors. Normalized vessels are more stable, less permeable, and have improved perfusion, leading to reduced hypoxia and enhanced drug delivery. However, vascular normalization is a transient process, and tumors can eventually adapt to overcome the effects of anti-angiogenic therapy.

  • Lymphangiogenesis: Lymphangiogenesis, the formation of new lymphatic vessels, is also important for tumor metastasis. Lymphatic vessels provide a route for cancer cells to spread to regional lymph nodes and distant organs. VEGF-C and VEGF-D are the main drivers of lymphangiogenesis in cancer. Blocking VEGF-C/D signaling can inhibit lymphatic metastasis.

  • Hypoxia: Tumor hypoxia is a major driver of cancer progression. Hypoxic cancer cells are more resistant to radiation and chemotherapy. Hypoxia also induces the expression of hypoxia-inducible factor-1 (HIF-1), which activates the transcription of genes involved in angiogenesis, metastasis, and glucose metabolism. Targeting HIF-1 or its downstream targets is a potential therapeutic strategy for overcoming hypoxia-induced resistance.

Targeting the tumor vasculature has been a major focus of cancer therapy for many years. Anti-angiogenic therapies have shown some success in certain cancers, but they are not effective in all patients. Understanding the mechanisms of resistance to anti-angiogenic therapy and developing strategies to overcome resistance are major challenges.

2.4 Extracellular Matrix (ECM)

The extracellular matrix (ECM) is a complex network of proteins and polysaccharides that provides structural support to tissues and regulates cell behavior. In the TME, the ECM is often dysregulated, with increased deposition of collagen, fibronectin, and other ECM components. ECM remodeling plays a critical role in cancer progression, influencing tumor growth, invasion, metastasis, and drug resistance.

  • ECM Composition and Structure: The ECM is composed of a variety of proteins, including collagens, fibronectin, laminin, and proteoglycans. The specific composition and structure of the ECM vary depending on the tissue and the stage of cancer development. In general, tumors have increased levels of collagen and fibronectin compared to normal tissues. The ECM can also be cross-linked by enzymes such as lysyl oxidase (LOX), making it more rigid and resistant to degradation.

  • ECM Remodeling: ECM remodeling is a dynamic process that involves the synthesis, degradation, and modification of ECM components. Matrix metalloproteinases (MMPs) are a family of enzymes that degrade ECM proteins. MMPs play a complex role in cancer progression, with some MMPs promoting tumor invasion and metastasis, while others inhibiting these processes. CAFs are a major source of MMPs in the TME.

  • Integrins: Integrins are transmembrane receptors that mediate cell-ECM interactions. They play a crucial role in cell adhesion, migration, and signaling. Integrins can activate downstream signaling pathways, such as the FAK and MAPK pathways, which promote cell survival and proliferation. Different integrins bind to different ECM components, and their expression patterns vary depending on the cancer type and stage. Targeting integrins has shown some promise in preclinical studies.

  • ECM and Drug Resistance: The ECM can contribute to drug resistance by sequestering drugs, inhibiting drug penetration, and activating survival pathways in cancer cells. The dense ECM can physically prevent drugs from reaching cancer cells. The ECM can also bind to drugs and reduce their bioavailability. Furthermore, ECM-integrin interactions can activate survival pathways that protect cancer cells from the cytotoxic effects of drugs.

Targeting the ECM has emerged as a promising therapeutic strategy for cancer. Strategies include inhibiting ECM synthesis, promoting ECM degradation, and blocking ECM-integrin interactions. However, the complexity of the ECM and its interactions with cancer cells present significant challenges.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3. The TME as a Driver of Intratumoral Heterogeneity

Intratumoral heterogeneity, the presence of diverse cancer cell populations within a single tumor, is a major obstacle to effective cancer therapy. The TME plays a crucial role in shaping intratumoral heterogeneity by creating selective pressures that drive the evolution of distinct cancer cell subpopulations. Different regions of the TME can have different nutrient availability, oxygen levels, and immune cell infiltrates, leading to the selection of cancer cells with different metabolic capacities, invasive potential, and drug sensitivities.

  • Spatial Heterogeneity: The TME exhibits significant spatial heterogeneity, with different regions of the tumor having different characteristics. For example, the tumor periphery may be more vascularized and have higher oxygen levels than the tumor core. The tumor-stroma interface may have a different immune cell composition than the tumor interior. This spatial heterogeneity can lead to the selection of cancer cells with different phenotypes and behaviors.

  • Metabolic Heterogeneity: Cancer cells can exhibit significant metabolic heterogeneity, with some cells relying on glycolysis for energy production, while others rely on oxidative phosphorylation. The TME can influence metabolic heterogeneity by altering nutrient availability and oxygen levels. For example, hypoxic regions of the tumor can select for cancer cells that are adapted to survive under low-oxygen conditions and that rely on glycolysis for energy production.

  • Epithelial-Mesenchymal Transition (EMT): EMT is a process by which epithelial cells lose their cell-cell adhesion and acquire a mesenchymal phenotype. EMT is associated with increased invasiveness, metastasis, and drug resistance. The TME can induce EMT by secreting growth factors and cytokines, such as TGF-β and EGF. CAFs can also promote EMT by remodeling the ECM and secreting MMPs.

  • Cancer Stem Cells (CSCs): Cancer stem cells (CSCs) are a subpopulation of cancer cells that have the ability to self-renew and differentiate into other cancer cell types. CSCs are thought to be responsible for tumor initiation, metastasis, and drug resistance. The TME can play a role in maintaining and expanding the CSC population. For example, hypoxic regions of the tumor can promote the survival and self-renewal of CSCs.

Understanding the mechanisms by which the TME drives intratumoral heterogeneity is crucial for developing effective personalized cancer therapies. Strategies that target the TME to reduce intratumoral heterogeneity may improve the efficacy of conventional cancer therapies.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4. Therapeutic Strategies Targeting the Tumor Microenvironment

Targeting the TME has emerged as a promising therapeutic strategy for cancer. Numerous approaches are being developed to disrupt pro-tumorigenic TME interactions, including strategies that remodel the ECM, modulate immune cell function, normalize tumor vasculature, and target CAFs.

4.1 ECM Remodeling

Strategies to remodel the ECM include inhibiting ECM synthesis, promoting ECM degradation, and blocking ECM-integrin interactions.

  • MMP Inhibitors: MMP inhibitors were initially developed to inhibit tumor invasion and metastasis by blocking the degradation of ECM proteins. However, clinical trials with broad-spectrum MMP inhibitors have been largely disappointing, likely due to their lack of specificity and off-target effects. More selective MMP inhibitors are being developed that target specific MMPs involved in tumor progression.

  • Lysyl Oxidase (LOX) Inhibitors: LOX is an enzyme that cross-links collagen fibers, making the ECM more rigid and resistant to degradation. LOX inhibitors can disrupt ECM cross-linking, making the ECM more pliable and facilitating drug penetration. LOX inhibitors are being investigated in preclinical and clinical studies.

  • Hyaluronidase: Hyaluronan is a major component of the ECM that can contribute to increased interstitial pressure and impaired drug delivery. Hyaluronidase is an enzyme that degrades hyaluronan. Preclinical studies have shown that hyaluronidase can improve drug delivery and enhance the efficacy of chemotherapy in certain cancers.

4.2 Immune Modulation

Strategies to modulate immune cell function include enhancing anti-tumor immunity and suppressing immunosuppression.

  • Immune Checkpoint Inhibitors: Immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 antibodies, have revolutionized cancer therapy by blocking inhibitory signaling pathways that suppress anti-tumor immunity. These therapies have shown remarkable efficacy in certain cancers, but they are not effective in all patients. Understanding the mechanisms of resistance to immune checkpoint blockade is a major focus of current research.

  • Adoptive Cell Therapy: Adoptive cell therapy involves the ex vivo expansion and activation of immune cells, followed by their infusion into the patient. CAR T-cell therapy, a type of adoptive cell therapy, has shown remarkable efficacy in hematologic malignancies. However, CAR T-cell therapy has been less effective in solid tumors, due in part to the immunosuppressive TME.

  • Cytokine Therapy: Cytokines, such as IL-2 and IFN-α, can stimulate anti-tumor immunity. IL-2 has been used to treat metastatic melanoma and renal cell carcinoma. IFN-α has been used to treat hairy cell leukemia and Kaposi’s sarcoma.

4.3 Vascular Normalization

Strategies to normalize tumor vasculature include targeting VEGF signaling and other pro-angiogenic factors.

  • Anti-VEGF Therapy: Anti-VEGF antibodies, such as bevacizumab, can block VEGF signaling and inhibit angiogenesis. Anti-VEGF therapy has shown some success in certain cancers, but it is not effective in all patients. Tumors can eventually adapt to overcome the effects of anti-VEGF therapy.

  • Tyrosine Kinase Inhibitors (TKIs): TKIs that target VEGF receptors can also inhibit angiogenesis. TKIs, such as sunitinib and sorafenib, have been used to treat renal cell carcinoma, hepatocellular carcinoma, and other cancers.

  • Angiopoietin-2 (Ang-2) Inhibitors: Ang-2 is another pro-angiogenic factor that promotes vessel destabilization and leakage. Ang-2 inhibitors are being developed as potential anti-angiogenic therapies.

4.4 Targeting CAFs

Strategies to target CAFs include inhibiting CAF activation, depleting CAFs, and reprogramming CAFs.

  • FAP Inhibitors: FAP is a protease that is highly expressed on CAFs. FAP inhibitors can block CAF activation and reduce ECM remodeling.

  • TGF-β Inhibitors: TGF-β is an immunosuppressive cytokine that is secreted by CAFs. TGF-β inhibitors can enhance anti-tumor immunity and reduce tumor growth.

  • Reprogramming CAFs: Strategies to reprogram CAFs include converting them into quiescent fibroblasts or inducing them to undergo apoptosis. Reprogramming CAFs may reduce their pro-tumorigenic effects and enhance the efficacy of other cancer therapies.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5. Challenges and Future Directions

Despite significant progress in understanding the role of the TME in cancer progression, several challenges remain in translating TME-targeted therapies into clinical success. One of the major challenges is the inherent complexity and variability of the TME across different cancer types and stages. The TME is not a static entity; it is a dynamic ecosystem that evolves over time in response to various factors, including cancer cell mutations, therapy, and the host immune response. This dynamic nature makes it difficult to predict how the TME will respond to targeted therapies. Furthermore, the TME is highly heterogeneous, with different regions of the tumor having different characteristics. This heterogeneity poses a challenge for developing effective TME-targeted therapies that can reach all regions of the tumor.

Another challenge is the lack of reliable biomarkers to identify patients who are most likely to benefit from TME-targeted therapies. Biomarkers that can predict the response to these therapies are urgently needed. These biomarkers could be based on the expression of specific TME components, such as CAFs, immune cells, or ECM proteins. They could also be based on genomic or proteomic profiling of the TME.

Future research should focus on developing more personalized approaches to targeting the TME. This requires a deeper understanding of the specific TME components and their interactions in different cancer contexts. It also requires the development of novel technologies to characterize the TME at the single-cell level. These technologies include single-cell RNA sequencing, spatial transcriptomics, and multiplex immunohistochemistry. These technologies can provide valuable insights into the cellular composition, spatial organization, and functional state of the TME.

Another promising area of research is the development of combination therapies that target both cancer cells and the TME. Combining TME-targeted therapies with conventional chemotherapy or immunotherapy may improve the efficacy of these therapies and overcome resistance. For example, combining anti-angiogenic therapy with chemotherapy can improve drug delivery and reduce tumor hypoxia. Combining immune checkpoint inhibitors with TME-targeted therapies can enhance anti-tumor immunity and overcome immune evasion.

In conclusion, the TME plays a critical role in cancer progression, metastasis, and therapeutic resistance. Targeting the TME has emerged as a promising therapeutic strategy for cancer. However, the complexity and variability of the TME present significant challenges. Future research should focus on developing more personalized approaches to targeting the TME and on developing combination therapies that target both cancer cells and the TME. A deeper understanding of the TME will pave the way for the development of more effective and personalized cancer therapies.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

References

  1. Hanahan, D., & Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell, 144(5), 646-674.
  2. Quail, D. F., & Joyce, J. A. (2017). Microenvironmental regulation of cancer. Nature medicine, 19(11), 1423-1437.
  3. Hinshaw, D. C., & Shevde, L. A. (2019). The tumor microenvironment intimately affects cancer stem cell plasticity and heterogeneity. Seminars in Cancer Biology, 54, 25-35.
  4. Sahai, E., Astsaturov, I., Cukierman, E., De Almeida, C. J., Gómez-Moreno, C., Kissil, J. L., … & Weinberg, R. A. (2020). A framework for advancing our understanding of cancer-associated fibroblasts. Nature Reviews Cancer, 20(3), 174-186.
  5. Joyce, J. A., & Fearon, D. T. (2015). T cell exclusion, immune privilege, and the tumor microenvironment. Science, 348(6230), 74-80.
  6. Carmeliet, P., & Jain, R. K. (2011). Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nature Reviews Drug Discovery, 10(6), 417-427.
  7. Lu, P., Weaver, V. M., & Werb, Z. (2012). The extracellular matrix: a dynamic niche in cancer progression. Journal of Cell Biology, 196(4), 395-406.
  8. Marusyk, A., Almendro, V., & Polyak, K. (2012). Intra-tumour heterogeneity: a driving force in cancer progression. Cancer discovery, 2(12), 1077-1082.
  9. De Palma, M., & Hanahan, D. (2012). The biology of personalized cancer medicine: targeting the microenvironment. Cell, 148(6), 1081-1086.
  10. Roma-Rodrigues, C., Espinoza, J. A., Fernandes, R., & Reis, F. (2019). Metformin revisited: an old drug with new promise in cancer therapy. Cancers, 11(12), 1901.
  11. Jiang, H., Hegde, A., DeNardo, D. G. (2017). Tumor-associated macrophages as potential therapeutic targets. Trends in Cancer. 3(11):717-739.
  12. Arneth, B. M., Frese, K. K., Palmieri, D., & Weigelt, B. (2023). Cancer cell-CAF communication in drug resistance. Trends in Cell Biology, 33(6), 506-522.
  13. Pickup, M. W., Mouw, J. K., & Weaver, V. M. (2014). The extracellular matrix modulates the hallmarks of cancer. EMBO Reports, 15(12), 1243-1253.

4 Comments

  1. The discussion on ECM remodeling highlights the complex and sometimes contradictory roles of MMPs. Could personalized medicine approaches, guided by detailed ECM profiling, help us leverage these enzymes for targeted cancer therapy, promoting localized invasion only when beneficial?

    • That’s a great point! ECM profiling could be a game-changer. By understanding the specific ECM landscape in each patient, we might be able to design therapies that selectively target MMPs. It could allow for promoting localized invasion to enhance drug delivery or immune cell infiltration only when it benefits treatment. It would require very precise control!

      Editor: MedTechNews.Uk

      Thank you to our Sponsor Esdebe

  2. The mention of spatial heterogeneity within the TME raises interesting questions about therapeutic delivery. Could nanoparticles or other targeted delivery systems be engineered to navigate these varying microenvironments and ensure effective drug distribution throughout the tumor?

    • That’s a fantastic point! Exploring nanoparticle engineering to navigate the TME’s spatial heterogeneity is crucial. Perhaps combining targeted delivery with stimuli-responsive nanoparticles could enhance drug distribution in specific microenvironments. This could involve engineering nanoparticles to respond to pH gradients or enzymatic activity that are unique to a specific spatial location within the TME.

      Editor: MedTechNews.Uk

      Thank you to our Sponsor Esdebe

Leave a Reply to MedTechNews.Uk Cancel reply

Your email address will not be published.


*