GET THE APP

Understanding Tumor-Associated Stroma: A Crucial Player in Cancer Progression and Therapy
..

Journal of Oncology Translational Research

ISSN: 2476-2261

Open Access

Perspective - (2023) Volume 9, Issue 1

Understanding Tumor-Associated Stroma: A Crucial Player in Cancer Progression and Therapy

Angelo Michael*
*Correspondence: Angelo Michael, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA, Email:
Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA

Received: 01-Feb-2023, Manuscript No. jotr-23-101644; Editor assigned: 03-Feb-2023, Pre QC No. P-101644; Reviewed: 15-Feb-2023, QC No. 101644; Revised: 21-Feb-2023, Manuscript No. R-101644; Published: 28-Feb-2023 , DOI: 10.37421/2476-2261.2023.9.221
Citation: Michael, Angelo. “Understanding Tumor-Associated Stroma: A Crucial Player in Cancer Progression and Therapy.” J Surg Path Diag 9 (2023): 221.
Copyright: © 2023 Michael A. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

Cancer is a complex disease characterized by the uncontrolled growth and spread of abnormal cells within the body. While cancer cells themselves play a critical role in tumor initiation and progression, emerging research has shed light on the importance of the tumor microenvironment, specifically the tumor-associated stroma. The tumor-associated stroma comprises various non-cancerous cells, Extracellular Matrix (ECM) components, and signalling molecules present within and around the tumor. This dynamic and complex network of interactions between cancer cells and the surrounding stroma significantly influences tumor behavior, response to therapy, and patient outcomes. In this article, we will explore the role of tumor-associated stroma in cancer development, progression, and therapeutic approaches. Tumor-associated stroma encompasses a diverse range of cellular and non-cellular components that interact with cancer cells to create a supportive microenvironment for tumor growth and metastasis. It consists of fibroblasts, immune cells (such as macrophages and lymphocytes), endothelial cells, pericytes, adipocytes, and various ECM components [1].

These components work in tandem to promote tumor progression through direct cell-cell interactions and secreted factors. Tumor-stroma interactions are bidirectional and dynamic. Cancer cells can induce changes in the stroma through the release of cytokines, growth factors, and proteases, leading to the activation of fibroblasts, recruitment of immune cells, and remodeling of the ECM. In turn, the stroma influences tumor behavior by providing growthpromoting signals, promoting angiogenesis, suppressing immune surveillance, and creating a physical barrier against drug penetration. Additionally, tumorassociated stroma can induce Epithelial-mesenchymal Transition (EMT), a process that enhances cancer cell invasiveness and metastatic potential. Tumor angiogenesis, the formation of new blood vessels to supply nutrients and oxygen to the growing tumor, is a crucial step in tumor progression. Tumor-associated stroma, particularly endothelial cells, pericytes, and fibroblasts, play a vital role in promoting angiogenesis through the secretion of angiogenic factors like Vascular Endothelial Growth Factor (VEGF) and Matrix Metalloproteinases (MMPs). Understanding these interactions has led to the development of anti-angiogenic therapies that target the tumor-associated stroma to disrupt tumor blood supply [2].

The immune system has the potential to recognize and eliminate cancer cells. However, tumors have evolved multiple strategies to evade immune surveillance. The tumor-associated stroma, including immune cells such as macrophages, regulatory T cells, and myeloid-derived suppressor cells, plays a significant role in creating an immunosuppressive environment. These cells secrete cytokines and chemokines that inhibit immune cell activation and promote an immunosuppressive phenotype. Targeting these interactions holds promise for immunotherapeutic interventions to enhance anti-tumor immune responses. Tumor-associated stroma has been implicated in the development of resistance to conventional cancer therapies, including chemotherapy and radiation. The stroma can act as a physical barrier, limiting drug delivery to the tumor cells. Moreover, the altered signalling pathways and increased production of growth factors by the stroma can activate survival mechanisms in cancer cells, leading to therapy resistance. Understanding these interactions is crucial for the development of strategies to overcome therapeutic resistance and improve patient outcomes [3].

Description

The unique characteristics and importance of tumor-associated stroma in cancer progression have sparked interest in targeting this microenvironment for therapeutic intervention. Various approaches are being explored; including stroma-modulating agents, anti-angiogenic therapies, immunotherapies, and combination treatments that aim to disrupt tumor-stroma interactions and enhance the efficacy of standard cancer treatments. The tumor-associated stroma is a critical component of the tumor microenvironment that significantly influences cancer development, progression, and response to therapy. Understanding the complex interactions between cancer cells and the stroma is essential for the development of effective treatment strategies. Targeting the tumor-associated stroma holds promise for improving patient outcomes and overcoming therapeutic resistance in cancer. Further research and clinical trials are needed to unravel the intricate mechanisms underlying tumor-stroma interactions and translate this knowledge into innovative therapies for cancer patients. While significant progress has been made in unraveling the role of tumor-associated stroma in cancer; there are several challenges that need to be addressed for effective targeting of this microenvironment. First, the heterogeneity of tumor-associated stroma across different cancer types and even within the same tumor poses a challenge in developing universal therapeutic strategies. Tailoring treatments to specific tumor-stroma characteristics and patient profiles will be crucial for optimal outcomes [4].

Second, the dynamic nature of tumor-stroma interactions necessitates a comprehensive understanding of the temporal and spatial changes occurring within the tumor microenvironment. Longitudinal studies and advanced imaging techniques will be instrumental in capturing these dynamic changes and guiding treatment decisions. Third, the development of reliable biomarkers to assess the activation state and functional properties of the tumor-associated stroma is essential for patient stratification and monitoring treatment responses. Biomarkers that can predict therapeutic outcomes and identify patients most likely to benefit from stroma-targeted therapies will enhance treatment precision. Lastly, the design of therapeutic strategies targeting tumor-associated stroma needs to consider potential side effects and unintended consequences. Disrupting the stroma could have adverse effects on normal tissue homeostasis and wound healing processes. Therefore, careful evaluation of the balance between therapeutic efficacy and potential toxicity is necessary.

Addressing the challenges associated with tumor-associated stroma requires a multidisciplinary and collaborative approach. Scientists, clinicians, and industry partners need to work together to advance our understanding of stromal biology, develop innovative therapeutic approaches, and translate these discoveries into clinical practice. Translational research, incorporating findings from preclinical models and clinical trials, will be crucial for identifying effective stroma-targeted therapies and optimizing their use in cancer patients [5].

Conclusion

In recent years, several therapeutic strategies targeting tumor-associated stroma have been explored in preclinical and clinical settings. One approach involves directly targeting the activated fibroblasts, also known as Cancer- Associated Fibroblasts (CAFs), which are key components of the tumor associated stroma. Various drugs and agents have been developed to specifically target CAFs, including small molecule inhibitors, monoclonal antibodies, and nanoparticle-based delivery systems. These agents aim to disrupt the protumorigenic functions of CAFs, such as ECM remodeling, angiogenesis promotion, and immune evasion. Another strategy focuses on targeting angiogenesis, a process tightly regulated by the tumor-associated stroma. Anti-angiogenic therapies, such as monoclonal antibodies against VEGF or its receptors, have been approved for the treatment of several cancer types. These agents inhibit the formation of new blood vessels within the tumor, thereby reducing nutrient supply and impeding tumor growth. However, resistance to anti-angiogenic therapies can occur, highlighting the need for further research to identify optimal treatment combinations and overcome resistance mechanisms.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Rivenbark, Ashley G., Siobhan M. O’Connor and William B. Coleman. "Molecular and cellular heterogeneity in breast cancer: Challenges for personalized medicine." Am J Clin Pathol 183 (2013): 1113-1124.
  2. Google Scholar, Crossref, Indexed at

  3. Pandey, Puspa Raj, Jamila Saidou and Kounosuke Watabe. "Role of myoepithelial cells in breast tumor progression." Front Biosci 15 (2010): 226.
  4. Google Scholar, Crossref, Indexed at

  5. Aumailley, Monique, Leena Bruckner-Tuderman, William G. Carter and Rainer Deutzmann, et al. "A simplified laminin nomenclature." Matrix Biol 24 (2005): 326-332.
  6. Google Scholar, Crossref, Indexed at

  7. Jayadev, Ranjay and David R. Sherwood. "Basement membranes." Curr 27 (2017): R207-R211.
  8. Google Scholar, Crossref

  9. Paavolainen, Oona and Emilia Peuhu. "Integrin-mediated adhesion and mechanosensing in the mammary gland." Semin Cell Dev Biol 114 (2021):113-125.
  10. Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward