Appearance
This report is written by MaltSci based on the latest literature and research findings
What is the role of tumor microenvironment in cancer progression?
Abstract
The tumor microenvironment (TME) is a pivotal aspect of cancer biology that significantly influences tumor progression and therapeutic outcomes. Comprising a diverse array of cellular and non-cellular components, the TME interacts dynamically with tumor cells, shaping their behavior and promoting processes such as proliferation, invasion, and metastasis. This review synthesizes current knowledge on the multifaceted roles of the TME in cancer dynamics, highlighting the contributions of various cellular components, including cancer-associated fibroblasts (CAFs), immune cells, and endothelial cells. Additionally, the TME's non-cellular components, such as the extracellular matrix (ECM) and secreted factors, play critical roles in facilitating tumor growth and immune evasion. The TME not only supports tumor development but also contributes to therapeutic resistance, complicating treatment strategies. By elucidating the complex interactions within the TME, this review aims to identify potential therapeutic targets that could enhance treatment efficacy and address challenges like drug resistance and immune suppression. Ultimately, understanding the TME offers novel avenues for innovative therapeutic interventions that may improve clinical outcomes for cancer patients.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Components of the Tumor Microenvironment
- 2.1 Cellular Components
- 2.2 Non-Cellular Components
- 3 The Role of TME in Tumor Growth and Metastasis
- 3.1 Tumor Cell Proliferation
- 3.2 Invasion and Metastatic Spread
- 4 TME and Immune Evasion
- 4.1 Immune Cell Interactions
- 4.2 Mechanisms of Immune Suppression
- 5 Angiogenesis in the Tumor Microenvironment
- 5.1 Vascularization Processes
- 5.2 Impact on Tumor Growth
- 6 Resistance to Therapy in the Context of TME
- 6.1 Mechanisms of Resistance
- 6.2 Therapeutic Implications
- 7 Conclusion
1 Introduction
The tumor microenvironment (TME) has emerged as a critical component in the study of cancer progression, encompassing a complex network of cellular and non-cellular elements that interact dynamically with tumor cells. Historically, cancer research has primarily focused on the intrinsic properties of malignant cells, such as genetic mutations and oncogene activation. However, it is now widely recognized that the TME significantly influences tumor behavior, including growth, metastasis, and therapeutic response [1][2]. The TME comprises various cell types, including stromal cells, immune cells, and endothelial cells, as well as extracellular matrix (ECM) components and soluble factors that facilitate intricate signaling pathways [3][4].
Understanding the role of the TME in cancer progression is of paramount importance, as it presents novel therapeutic opportunities that extend beyond traditional cytotoxic approaches. By elucidating the interactions between tumor cells and their surrounding environment, researchers can identify potential targets for intervention that may enhance treatment efficacy and overcome challenges such as drug resistance and immune evasion [5][6]. The TME not only supports tumor growth but also plays a pivotal role in shaping the metastatic potential of cancer cells, making it a focal point for therapeutic strategies aimed at disrupting these interactions [7][8].
Current research has highlighted several key aspects of the TME that are critical to its role in cancer dynamics. These include the cellular components of the TME, such as cancer-associated fibroblasts (CAFs), immune cells, and adipocytes, which contribute to the tumor's supportive niche [9][10]. Additionally, the non-cellular components, including the ECM and various signaling molecules, create a microenvironment that not only sustains tumor growth but also facilitates angiogenesis and immune suppression [7][11].
This review is structured to provide a comprehensive overview of the multifaceted roles of the TME in cancer progression. The first section will detail the components of the TME, distinguishing between cellular and non-cellular elements and their respective contributions to tumor biology. Following this, we will explore the TME's role in tumor growth and metastasis, focusing on mechanisms such as tumor cell proliferation and invasive behavior. We will then examine how the TME facilitates immune evasion, detailing the interactions between tumor cells and immune components, as well as the mechanisms underlying immune suppression. The subsequent section will address angiogenesis within the TME, discussing the processes involved in vascularization and their implications for tumor growth. Finally, we will analyze the relationship between the TME and resistance to therapy, exploring how the microenvironment contributes to therapeutic failure and outlining potential strategies for overcoming these challenges.
In conclusion, the TME represents a dynamic and integral aspect of cancer biology that significantly impacts tumor progression and treatment outcomes. By synthesizing current findings, this review aims to provide insights into the complex interplay between tumor cells and their microenvironment, ultimately identifying avenues for innovative therapeutic interventions that target the TME to improve clinical outcomes for cancer patients.
2 Components of the Tumor Microenvironment
2.1 Cellular Components
The tumor microenvironment (TME) plays a critical role in cancer progression through its complex composition and dynamic interactions with cancer cells. It consists of various cellular components, including fibroblasts, adipocytes, endothelial cells, immune cells, cancer stem cells, and the extracellular matrix (ECM). These components create a supportive niche that not only fosters tumor growth but also influences cancer cell behavior, including proliferation, invasion, and resistance to therapies.
Fibroblasts, as a major component of the TME, contribute to the structural integrity of the tumor by producing ECM proteins and cytokines that facilitate tumor growth and survival. They also participate in the remodeling of the ECM, which can alter the physical properties of the tumor and promote invasion and metastasis (Franco et al., 2020; Sounni & Noel, 2013). Immune cells, including tumor-associated macrophages and T cells, can have dual roles, either promoting tumor progression by creating an immunosuppressive environment or inhibiting it through immune-mediated mechanisms. This complexity underscores the need to understand the interplay between these cells and their contributions to tumor biology (Turlej et al., 2025; Di Ceglie et al., 2024).
The ECM itself, composed of various proteins and soluble factors, serves not only as a scaffold for tumor cells but also as a mediator of signaling pathways that regulate cell behavior. Changes in ECM composition and stiffness can lead to enhanced tumor cell migration and invasion, facilitating metastasis (González-González & Alonso, 2018). Additionally, the TME is involved in angiogenesis, providing necessary nutrients and oxygen to the tumor, further supporting its growth (Kharouf et al., 2023).
Moreover, the TME is implicated in the development of drug resistance. Cancer cells often exploit the protective niche provided by the TME to evade the effects of chemotherapy and targeted therapies, leading to treatment failure (Paraiso & Smalley, 2013). This highlights the importance of targeting both cancer cells and the TME in therapeutic strategies to improve clinical outcomes.
In summary, the tumor microenvironment, through its diverse cellular components and their interactions, plays a pivotal role in facilitating cancer progression by supporting tumor growth, promoting invasion and metastasis, and contributing to therapeutic resistance. Understanding these interactions is crucial for the development of more effective cancer therapies.
2.2 Non-Cellular Components
The tumor microenvironment (TME) plays a crucial role in cancer progression, acting as a dynamic and complex system that influences various aspects of tumor biology. The TME comprises both cellular and non-cellular components, with the latter including a variety of secreted factors that significantly contribute to cancer survival, progression, and drug resistance.
Non-cellular components of the TME consist of various secreted factors such as growth factors, cytokines, RNA, DNA, metabolites, and structural matrix proteins. These elements are vital in orchestrating a multitude of processes that support cancer cells. For instance, they provide essential metabolites and energy, deliver growth signals, facilitate immune evasion, create environments conducive to drug resistance, and promote metastatic and angiogenic cues (Patel et al., 2018) [12].
Moreover, the interplay between tumor cells and their surrounding environment is essential for the manifestation of malignant features. The TME influences tumor cell behaviors, including proliferation, migration, and the ability to metastasize, through complex interactions with immune cells, fibroblasts, and the extracellular matrix (Sounni & Noel, 2013) [6]. The non-cellular components of the TME, such as the extracellular matrix (ECM), also play a pivotal role by modulating the signaling pathways that govern tumor behavior, highlighting the necessity of understanding these interactions for therapeutic advancements (Xiao & Yu, 2021) [13].
Matricellular proteins, a subgroup of non-cellular components, have gained attention for their multifaceted roles in cancer development and progression. These proteins, which include periostin and osteopontin, are involved in critical aspects of tumor biology, such as matrix remodeling, invasion, and the establishment of pre-metastatic niches (González-González & Alonso, 2018) [9]. Their production can be influenced by both tumor cells and tumor-associated cells, making them significant targets for therapeutic intervention.
Furthermore, the non-cellular components of the TME can facilitate a shift from tumor-suppressive to tumor-promoting environments. For instance, during the early stages of tumor development, the TME may exhibit suppressive characteristics, but as the tumor progresses, it can transform into a microenvironment that promotes immune tolerance and tumor cell malignancy (Liu et al., 2022) [14]. This transformation underscores the importance of targeting these non-cellular components to disrupt the supportive environment that tumors exploit for growth and metastasis.
In summary, the non-cellular components of the tumor microenvironment are integral to cancer progression, influencing tumor cell behavior and the overall tumor landscape. Understanding these components and their interactions with tumor cells is critical for developing effective therapeutic strategies aimed at disrupting the supportive roles they play in cancer biology.
3 The Role of TME in Tumor Growth and Metastasis
3.1 Tumor Cell Proliferation
The tumor microenvironment (TME) plays a critical role in cancer progression, influencing various aspects of tumor cell behavior, including proliferation, invasion, and metastasis. The TME consists of a complex network of cellular and non-cellular components, including stromal cells, extracellular matrix (ECM), signaling molecules, and immune cells, all of which interact dynamically with cancer cells.
Cancer cells do not operate in isolation; rather, they are deeply embedded in the TME, which significantly impacts their growth and metastatic potential. The interactions between cancer cells and their surrounding environment are fundamental to the tumor's development and progression. For instance, the TME provides essential growth signals and metabolic support to cancer cells. Tumor cells actively recruit stromal cells, which in turn secrete growth factors, cytokines, and other signaling molecules that facilitate tumor growth and survival [1].
One of the mechanisms by which the TME influences tumor cell proliferation is through the remodeling of the ECM. The ECM not only serves as a structural scaffold but also participates in biochemical signaling that regulates cell behavior. Changes in the composition and architecture of the ECM can promote tumor cell proliferation and invasion by altering the mechanical properties of the microenvironment, which is critical for tumor progression [15]. The mechanical stiffness of the TME, for example, has been shown to affect cancer cell migration and invasion, creating a feedback loop that enhances tumor aggressiveness [15].
Moreover, the presence of immune cells within the TME, particularly tumor-associated macrophages and myeloid-derived suppressor cells, can also drive tumor cell proliferation. These immune cells can create an immunosuppressive environment that allows cancer cells to evade immune surveillance, thereby promoting their growth and metastatic potential [16]. The reciprocal interactions between cancer cells and immune cells in the TME are pivotal, as they can lead to increased proliferation and survival of tumor cells, contributing to the overall aggressiveness of the cancer [17].
In addition to promoting tumor growth, the TME also plays a crucial role in metastasis. It influences the process of epithelial-mesenchymal transition (EMT), a critical step that allows cancer cells to detach from the primary tumor, invade surrounding tissues, and disseminate to distant sites [5]. The TME's composition, including the presence of specific cell types and the biochemical signals they produce, can enhance the invasive capabilities of cancer cells, thereby facilitating metastasis [18].
In summary, the tumor microenvironment is not merely a passive background but an active participant in cancer progression. It shapes tumor cell behavior through a myriad of interactions that promote proliferation, survival, and metastatic potential. Understanding these interactions provides insights into potential therapeutic strategies aimed at targeting the TME to inhibit cancer progression and improve treatment outcomes.
3.2 Invasion and Metastatic Spread
The tumor microenvironment (TME) plays a critical role in cancer progression, particularly in the processes of invasion and metastatic spread. It consists of various cellular components, including stromal cells, immune cells, and the extracellular matrix (ECM), which interact dynamically with cancer cells, influencing their behavior and fate.
Cancer progression is inherently linked to the TME, as it is not merely a passive backdrop but an active participant in tumorigenesis. Tumor cells recruit stromal cells that provide essential growth signals, intermediate metabolites, and a supportive environment conducive to tumor growth and metastasis [1]. This reciprocal communication between tumor cells and the TME enhances their proliferative and invasive capabilities, allowing cancer cells to thrive and spread.
Invasion is a crucial step in the metastatic process, where cancer cells must penetrate the surrounding tissues. The TME facilitates this invasion through several mechanisms. For instance, cancer cells can alter the composition and structure of the ECM, which affects their motility and the ability to migrate through tissues [19]. The formation of invadopodia, actin-rich structures that degrade ECM components, is one such mechanism that enables cancer cells to invade neighboring tissues [19].
Moreover, the TME influences the metastatic spread of cancer cells to distant organs. The creation of a metastatic niche—an environment that supports the survival and proliferation of disseminated tumor cells—is critical for successful metastasis [20]. This niche is shaped by interactions between cancer cells and the host stroma, which can include the secretion of growth factors, cytokines, and other signaling molecules that facilitate the colonization of secondary sites [16]. The dynamics of this microenvironment can also involve changes in immune cell populations, such as the recruitment of myeloid-derived suppressor cells (MDSCs), which promote immune tolerance and further support tumor growth and metastasis [16].
Inflammation within the TME is another significant factor that drives cancer metastasis. Proinflammatory cytokines and chemokines can enhance the invasive properties of cancer cells and promote angiogenesis, which is vital for supplying nutrients to growing tumors [21]. This inflammatory milieu not only fosters tumor proliferation but also contributes to the establishment of a supportive environment for metastasis [21].
In summary, the tumor microenvironment is integral to cancer progression, particularly in invasion and metastatic spread. It actively shapes the behavior of cancer cells through various cellular and molecular interactions, creating conditions that favor tumor growth and dissemination. Understanding these interactions offers potential therapeutic targets for interrupting the processes of invasion and metastasis, thereby improving cancer treatment outcomes [5][18][22].
4 TME and Immune Evasion
4.1 Immune Cell Interactions
The tumor microenvironment (TME) plays a critical role in cancer progression by creating a complex ecosystem that not only supports tumor growth but also facilitates immune evasion. The TME consists of various cellular and non-cellular components, including cancer cells, stromal cells, immune cells, and extracellular matrix (ECM), which interact dynamically to influence tumor behavior and the immune response.
Immune cells within the TME can have both pro-tumor and anti-tumor effects, depending on their type and activation state. For instance, myeloid-derived suppressor cells (MDSCs) are a significant component of the TME that contribute to immunosuppression, promoting tumor growth and metastasis by inhibiting the activity of T cells and other immune cells. MDSCs disrupt antitumor T cell activity and are often found in elevated numbers in cancer patients, where they play a key role in creating an immunosuppressive microenvironment [23].
The interaction between immune cells and tumor cells is further complicated by the presence of regulatory T cells (Tregs), which can suppress immune responses against tumors, leading to immune evasion. Tumors can evade immune surveillance through various mechanisms, including downregulating antigen presentation and upregulating immune checkpoint molecules, which inhibit T cell activation [24].
Moreover, the extracellular matrix (ECM) and soluble factors in the TME also influence immune cell behavior. The ECM can modulate the mechanical properties of the tumor environment, affecting how immune cells migrate and interact with tumor cells. For example, the stiffness of the ECM can impact T cell infiltration and activation [25]. Additionally, soluble factors such as cytokines and chemokines secreted by tumor cells and stromal cells can create a local environment that either attracts or repels immune cells, thereby influencing the overall immune landscape [26].
Furthermore, hypoxia, a common feature of the TME, plays a significant role in promoting immune evasion. Hypoxic conditions can lead to the upregulation of immune checkpoint molecules and other factors that suppress immune responses, enabling cancer cells to thrive despite the presence of immune cells [27].
The interplay between different immune cell types, such as macrophages and neutrophils, is also crucial in shaping the TME. These cells can switch between pro-inflammatory and anti-inflammatory states, influenced by signals from tumor cells and the ECM. This plasticity is pivotal in determining whether the immune response will be effective against the tumor or contribute to its progression [28].
In summary, the TME significantly influences cancer progression through its complex interactions with immune cells. By fostering an immunosuppressive environment, the TME not only supports tumor growth but also enables cancer cells to evade immune surveillance, complicating treatment strategies and underscoring the need for therapies that target both tumor cells and their microenvironment [3][29][30].
4.2 Mechanisms of Immune Suppression
The tumor microenvironment (TME) plays a pivotal role in cancer progression and immune evasion through a complex interplay of various cellular and non-cellular components. The TME is characterized by a heterogeneous mix of cancer cells, immune cells, fibroblasts, endothelial cells, and the extracellular matrix, all of which contribute to tumor growth and the modulation of immune responses.
One of the primary mechanisms by which the TME facilitates immune evasion is through the suppression of anti-tumor immune responses. Cancer cells can exploit intrinsic mechanisms to dampen host immune activity, such as upregulating immune checkpoints, impairing antigen presentation, and competing for essential nutrients. For instance, immune checkpoint molecules like PD-L1 are often overexpressed by tumor cells and can inhibit T cell function, thereby promoting tumor immune escape [31].
Additionally, the TME can induce a state of immune tolerance, wherein the immune system is unable to mount an effective response against tumor cells. This phenomenon is often facilitated by myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), which are recruited to the TME and can suppress the activation and proliferation of effector T cells. MDSCs, in particular, are known to produce immunosuppressive cytokines and can inhibit T cell function directly, thereby enhancing tumor survival and progression [16].
The physical and biochemical properties of the TME, such as hypoxia, also contribute significantly to immune suppression. Hypoxic conditions can lead to the activation of various signaling pathways that reshape the immune microenvironment, resulting in an immunosuppressive state that favors tumor growth [32]. Moreover, the TME can produce soluble factors and exosomes that carry immune-modulatory molecules, further facilitating the suppression of anti-tumor immunity [31].
The dynamic nature of the TME means that it can evolve alongside tumor progression, continuously adapting to support cancer cell survival while inhibiting immune responses. For example, the interactions between cancer cells and the extracellular matrix can influence the behavior of immune cells within the TME, creating a feedback loop that perpetuates immune evasion and tumor growth [27].
In summary, the tumor microenvironment is a critical player in cancer progression, primarily through mechanisms of immune suppression that allow tumors to evade immune surveillance. Understanding these interactions is essential for developing targeted therapies aimed at manipulating the TME to enhance the immune system's ability to combat tumors [27][30][31].
5 Angiogenesis in the Tumor Microenvironment
5.1 Vascularization Processes
The tumor microenvironment (TME) plays a critical role in cancer progression, particularly through the processes of angiogenesis, which is the formation of new blood vessels within the tumor. This process is essential for providing the necessary nutrients and oxygen to tumor cells, thus facilitating their growth, invasion, and metastasis.
Angiogenesis in the TME is characterized by a complex interplay of various cellular components, including tumor cells, stromal cells, immune cells, and endothelial cells. The TME is influenced by both proangiogenic and antiangiogenic factors, creating an imbalance that favors tumor vascularization. This imbalance results in the formation of tortuous and hyperpermeable blood vessels, which not only support tumor growth but also contribute to a hostile microenvironment characterized by hypoxia and acidosis. Such conditions further stimulate angiogenesis and can lead to poor therapeutic outcomes [33].
The TME components, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and regulatory T cells (Treg), play pivotal roles in modulating angiogenesis. These stromal cells secrete various cytokines and growth factors that promote angiogenesis, thus creating a conducive environment for tumor progression [34].
Moreover, the abnormal metabolic conditions within the TME, such as low oxygen levels and acidic pH, can upregulate the expression of angiogenic factors, further driving the angiogenic process. High levels of inflammatory cytokines and angiogenic factors in the TME are associated with increased tumor growth and metastasis [35].
The normalization of the tumor microvasculature through targeted therapies that modulate the TME has emerged as a promising strategy for anti-angiogenesis and cancer treatment. By restoring the balance between pro- and anti-angiogenic factors, these therapies aim to improve the effectiveness of conventional treatments like chemotherapy and radiotherapy [33].
In summary, the tumor microenvironment is integral to the processes of angiogenesis and vascularization, significantly influencing cancer progression. The interplay between various cellular components and the metabolic conditions within the TME not only facilitates tumor growth but also presents opportunities for therapeutic interventions aimed at disrupting these processes [36] [37].
5.2 Impact on Tumor Growth
The tumor microenvironment (TME) plays a critical role in cancer progression, particularly through the process of angiogenesis, which is the formation of new blood vessels within the tumor. This process is essential for tumor growth and metastasis, as it ensures an adequate supply of oxygen and nutrients while facilitating the removal of metabolic waste. The TME is not merely a passive background; it actively contributes to tumor progression through complex interactions among various components, including tumor cells, immune cells, fibroblasts, and the extracellular matrix.
Angiogenesis within the TME is regulated by a balance of proangiogenic and antiangiogenic factors. Factors such as vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) are pivotal in promoting angiogenesis, while other factors can inhibit this process. The interplay between these factors determines the vascularization necessary for tumor growth. The presence of hypoxia within the TME often triggers the expression of proangiogenic factors, thereby enhancing the angiogenic response (Kaur & Roy, 2024)[36].
The composition of the TME varies across different tumor types, but common features include immune cells, stromal cells, and the extracellular matrix. Tumor cells induce significant molecular, cellular, and physical changes in their surrounding tissues, fostering an environment conducive to their own survival and proliferation. For instance, the activation of immune cells can lead to both pro- and anti-tumorigenic effects, complicating the role of the immune response in cancer progression (Anderson & Simon, 2020)[38].
Moreover, the TME can influence the efficacy of therapeutic interventions. Current anti-angiogenic therapies often face challenges due to the dynamic nature of the TME, which can adapt and create a hypoxic environment that limits drug effectiveness. High levels of inflammatory cytokines and angiogenic factors in the TME can also contribute to resistance against therapies aimed at inhibiting angiogenesis (Jiang et al., 2020)[35].
In summary, the tumor microenvironment is integral to cancer progression through its role in angiogenesis, impacting tumor growth and response to therapies. A comprehensive understanding of these interactions is essential for developing effective therapeutic strategies targeting the TME, thereby improving clinical outcomes for cancer patients.
6 Resistance to Therapy in the Context of TME
6.1 Mechanisms of Resistance
The tumor microenvironment (TME) plays a critical role in cancer progression and therapy resistance through its complex interactions with tumor cells. The TME consists of various cellular and non-cellular components, including stromal cells, extracellular matrix (ECM), soluble factors, and immune cells, which collectively influence tumor growth, survival, and response to treatment.
One of the primary mechanisms by which the TME contributes to therapy resistance is through the establishment of a supportive niche for cancer cells. For instance, cancer-associated fibroblasts (CAFs), a major component of the TME, create a paracrine environment that aids in the survival of tumor cells against therapeutic agents. These fibroblasts can secrete growth factors and cytokines that enhance tumor cell proliferation and contribute to the development of drug resistance [39]. Additionally, the TME can protect residual cancer cells from the cytotoxic effects of chemotherapy and radiation by providing physical barriers and biochemical signals that promote cell survival [40].
The immune cells present in the TME also play a significant role in modulating the response to therapy. For example, myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) can create an immunosuppressive environment that hinders the effectiveness of immunotherapies. MDSCs can inhibit T cell activation and promote tumor growth, while Tregs can suppress anti-tumor immune responses, leading to treatment failure [16].
Moreover, the TME influences the metabolic state of cancer cells, which can alter their sensitivity to therapies. Tumor cells often undergo metabolic reprogramming to adapt to the nutrient-depleted and hypoxic conditions of the TME, leading to enhanced survival and proliferation. This metabolic adaptation can contribute to the development of resistance against various therapeutic modalities [41].
Furthermore, the dynamic interactions between tumor cells and the TME can lead to the activation of signaling pathways associated with drug resistance. For instance, the Wnt/β-catenin signaling pathway has been implicated in promoting chemoresistance in colorectal cancer by facilitating the communication between tumor cells and the TME [42]. This cross-talk enables tumor cells to exploit the supportive features of the TME, ultimately leading to increased metastatic potential and therapeutic resistance [43].
In summary, the tumor microenvironment plays a multifaceted role in cancer progression and resistance to therapy by providing a supportive niche for tumor cells, modulating immune responses, influencing metabolic adaptations, and activating resistance-associated signaling pathways. Understanding these mechanisms is crucial for developing effective therapeutic strategies that target the TME and overcome the challenges of drug resistance.
6.2 Therapeutic Implications
The tumor microenvironment (TME) plays a crucial role in cancer progression and therapeutic resistance. It comprises a complex network of tumor cells, immune components, stromal cells, extracellular matrix (ECM), and various bioactive molecules, all of which interact dynamically to influence tumor behavior and response to therapy.
One of the primary functions of the TME is to provide physical support and biochemical signals that promote tumor development and survival. For instance, cancer-associated fibroblasts (CAFs) within the TME are instrumental in remodeling the ECM, facilitating angiogenesis, and modulating immune responses. These fibroblasts engage in cross-talk with cancer cells and immune cells through paracrine signaling, which can lead to immunosuppression and enhanced tumor cell proliferation [44]. The interactions between CAFs and other components of the TME significantly contribute to the resistance of tumors to therapies, including chemotherapy and radiotherapy [45].
Therapeutic resistance is often exacerbated by the TME's ability to create an immunosuppressive environment. For example, tumor-associated macrophages (TAMs) within the TME can secrete various factors that promote tumor growth and immune evasion. The secretory factors from both cancer cells and TAMs are crucial in modulating the tumor microenvironment, thereby influencing drug resistance [46]. Additionally, conditions such as hypoxia and altered metabolic states within the TME further complicate treatment responses by promoting metabolic adaptations that allow cancer cells to survive despite therapeutic interventions [47].
Given the TME's multifaceted role in fostering therapeutic resistance, targeting the TME has emerged as a promising strategy to enhance treatment efficacy. Current and emerging therapeutic strategies focus on disrupting the interactions within the TME, such as inhibiting CAF activity, reprogramming TAMs from an immunosuppressive to an immunostimulatory phenotype, and employing therapies that target the ECM [48]. Combination therapies that integrate TME-targeted agents with conventional treatments like chemotherapy and immunotherapy are being explored to improve treatment outcomes and overcome resistance mechanisms [47].
Moreover, innovative approaches, including the use of nanoparticles responsive to TME stimuli, are being developed to enhance drug delivery and therapeutic efficacy in the context of drug resistance [49]. Understanding the intricate interactions within the TME and their implications for therapy is essential for developing more effective, personalized cancer treatments.
In summary, the TME is a critical determinant of cancer progression and therapeutic resistance. Its complex interactions and components not only support tumor growth but also present significant challenges to effective treatment. Addressing the therapeutic implications of the TME through targeted interventions offers a promising avenue for improving patient outcomes in cancer therapy.
7 Conclusion
The tumor microenvironment (TME) is a crucial factor in cancer progression, influencing tumor behavior through its complex interplay of cellular and non-cellular components. Key findings highlight the roles of various cellular elements such as cancer-associated fibroblasts (CAFs), immune cells, and endothelial cells, which create a supportive niche for tumor growth, invasion, and metastasis. Additionally, the TME's non-cellular components, including the extracellular matrix (ECM) and soluble factors, are integral in modulating signaling pathways that dictate tumor dynamics. Current research indicates that the TME not only fosters tumor proliferation but also facilitates immune evasion and therapeutic resistance, underscoring its significance as a target for novel therapeutic strategies. As we look to the future, further exploration of the TME's role in shaping cancer progression is essential. Future research should focus on understanding the mechanisms of TME-induced drug resistance and developing therapies that can effectively disrupt these interactions, potentially leading to improved treatment outcomes. The integration of TME-targeted approaches with existing therapies may enhance their efficacy and overcome the challenges posed by the tumor microenvironment, ultimately benefiting cancer patients.
References
- [1] Yao Yuan;Yu-Chen Jiang;Chong-Kui Sun;Qian-Ming Chen. Role of the tumor microenvironment in tumor progression and the clinical applications (Review).. Oncology reports(IF=3.9). 2016. PMID:26986034. DOI: 10.3892/or.2016.4660.
- [2] Edgardo V Ariztia;Catherine J Lee;Radhika Gogoi;David A Fishman. The tumor microenvironment: key to early detection.. Critical reviews in clinical laboratory sciences(IF=5.5). 2006. PMID:17050079. DOI: 10.1080/10408360600778836.
- [3] Smita Kumari;Dia Advani;Sudhanshu Sharma;Rashmi K Ambasta;Pravir Kumar. Combinatorial therapy in tumor microenvironment: Where do we stand?. Biochimica et biophysica acta. Reviews on cancer(IF=8.3). 2021. PMID:34224836. DOI: 10.1016/j.bbcan.2021.188585.
- [4] Eliza Turlej;Aleksandra Domaradzka;Justyna Radzka;Dominika Drulis-Fajdasz;Julita Kulbacka;Agnieszka Gizak. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression.. Cells(IF=5.2). 2025. PMID:40136652. DOI: 10.3390/cells14060403.
- [5] Feng Gao;Bin Liang;Srinivasa T Reddy;Robin Farias-Eisner;Xiulan Su. Role of inflammation-associated microenvironment in tumorigenesis and metastasis.. Current cancer drug targets(IF=3.5). 2014. PMID:24200082. DOI: 10.2174/15680096113136660107.
- [6] Nor Eddine Sounni;Agnès Noel. Targeting the tumor microenvironment for cancer therapy.. Clinical chemistry(IF=6.3). 2013. PMID:23193058. DOI: 10.1373/clinchem.2012.185363.
- [7] James Mason;Daniel Öhlund. Key aspects for conception and construction of co-culture models of tumor-stroma interactions.. Frontiers in bioengineering and biotechnology(IF=4.8). 2023. PMID:37091337. DOI: 10.3389/fbioe.2023.1150764.
- [8] Ping Zhou;Simon L Lu;Liang Chang;Baoying Liao;Ming Cheng;Xiaolin Xu;Xin Sui;Fenting Liu;Mingshu Zhang;Yinxue Wang;Rui Yang;Rong Li;Heng Pan;Chao Zhang. The pan-cancer landscape of abnormal DNA methylation and intratumor microorganisms.. Neoplasia (New York, N.Y.)(IF=7.7). 2023. PMID:36791577. DOI: 10.1016/j.neo.2023.100882.
- [9] Laura González-González;Javier Alonso. Periostin: A Matricellular Protein With Multiple Functions in Cancer Development and Progression.. Frontiers in oncology(IF=3.3). 2018. PMID:29946533. DOI: 10.3389/fonc.2018.00225.
- [10] Luís Henrique Corrêa;Rafael Corrêa;Cecília Menezes Farinasso;Lívia Pimentel de Sant'Ana Dourado;Kelly Grace Magalhães. Adipocytes and Macrophages Interplay in the Orchestration of Tumor Microenvironment: New Implications in Cancer Progression.. Frontiers in immunology(IF=5.9). 2017. PMID:28970834. DOI: 10.3389/fimmu.2017.01129.
- [11] Christopher Fleming;Samantha Morrissey;Yihua Cai;Jun Yan. γδ T Cells: Unexpected Regulators of Cancer Development and Progression.. Trends in cancer(IF=17.5). 2017. PMID:28780933. DOI: 10.1016/j.trecan.2017.06.003.
- [12] Himadri Patel;Pritish Nilendu;Devashree Jahagirdar;Jayanta K Pal;Nilesh Kumar Sharma. Modulating secreted components of tumor microenvironment: A masterstroke in tumor therapeutics.. Cancer biology & therapy(IF=4.6). 2018. PMID:29219656. DOI: 10.1080/15384047.2017.1394538.
- [13] Yi Xiao;Dihua Yu. Tumor microenvironment as a therapeutic target in cancer.. Pharmacology & therapeutics(IF=12.5). 2021. PMID:33259885. DOI: 10.1016/j.pharmthera.2020.107753.
- [14] Yuanda Liu;Changfeng Li;Yaoping Lu;Chang Liu;Wei Yang. Tumor microenvironment-mediated immune tolerance in development and treatment of gastric cancer.. Frontiers in immunology(IF=5.9). 2022. PMID:36341377. DOI: 10.3389/fimmu.2022.1016817.
- [15] Bashar Emon;Jessica Bauer;Yasna Jain;Barbara Jung;Taher Saif. Biophysics of Tumor Microenvironment and Cancer Metastasis - A Mini Review.. Computational and structural biotechnology journal(IF=4.1). 2018. PMID:30128085. DOI: 10.1016/j.csbj.2018.07.003.
- [16] Zhongnan Yin;Chunxiao Li;Junjie Wang;Lixiang Xue. Myeloid-derived suppressor cells: Roles in the tumor microenvironment and tumor radiotherapy.. International journal of cancer(IF=4.7). 2019. PMID:29992569. DOI: 10.1002/ijc.31744.
- [17] Shubhangi Singh;Ajay P Singh;Ranjana Mitra. Cancer-Associated Fibroblasts: Major Co-Conspirators in Tumor Development.. Cancers(IF=4.4). 2024. PMID:38201638. DOI: 10.3390/cancers16010211.
- [18] Simon P Daoust;Lenore Fahrig;Amanda E Martin;Frédéric Thomas. From forest and agro-ecosystems to the microecosystems of the human body: what can landscape ecology tell us about tumor growth, metastasis, and treatment options?. Evolutionary applications(IF=3.2). 2013. PMID:23396712. DOI: 10.1111/eva.12031.
- [19] Christine M Gould;Sara A Courtneidge. Regulation of invadopodia by the tumor microenvironment.. Cell adhesion & migration(IF=3.5). 2014. PMID:24714597. DOI: 10.4161/cam.28346.
- [20] Arnaud Descot;Thordur Oskarsson. The molecular composition of the metastatic niche.. Experimental cell research(IF=3.5). 2013. PMID:23707205. DOI: .
- [21] Yadi Wu;Binhua P Zhou. Inflammation: a driving force speeds cancer metastasis.. Cell cycle (Georgetown, Tex.)(IF=3.4). 2009. PMID:19770594. DOI: 10.4161/cc.8.20.9699.
- [22] J Thomas Parsons;Jill Slack-Davis;Robert Tilghman;W Gregory Roberts. Focal adhesion kinase: targeting adhesion signaling pathways for therapeutic intervention.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2008. PMID:18245520. DOI: 10.1158/1078-0432.CCR-07-2220.
- [23] Anik Pramanik;Sankar Bhattacharyya. Myeloid derived suppressor cells and innate immune system interaction in tumor microenvironment.. Life sciences(IF=5.1). 2022. PMID:35780842. DOI: 10.1016/j.lfs.2022.120755.
- [24] Nikita Mundhara;Pritam Sadhukhan. Cracking the Codes behind Cancer Cells' Immune Evasion.. International journal of molecular sciences(IF=4.9). 2024. PMID:39201585. DOI: 10.3390/ijms25168899.
- [25] Shoham Benmelech;Thien Le;Maggie McKay;Jungmin Nam;Krupakar Subramaniam;Daniela Tellez;Grace Vlasak;Michael Mak. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions.. APL bioengineering(IF=4.1). 2024. PMID:38572312. DOI: 10.1063/5.0195244.
- [26] Dolores Aguilar-Cazares;Rodolfo Chavez-Dominguez;Mario Marroquin-Muciño;Mario Perez-Medina;Jesus J Benito-Lopez;Angel Camarena;Uriel Rumbo-Nava;Jose S Lopez-Gonzalez. The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment.. Frontiers in endocrinology(IF=4.6). 2022. PMID:36072935. DOI: 10.3389/fendo.2022.929572.
- [27] Hongyang Yu;Jinyang Li;Shiyin Peng;Qin Liu;Dongfeng Chen;Zongsheng He;Junyu Xiang;Bin Wang. Tumor microenvironment: Nurturing cancer cells for immunoevasion and druggable vulnerabilities for cancer immunotherapy.. Cancer letters(IF=10.1). 2024. PMID:39645024. DOI: 10.1016/j.canlet.2024.217385.
- [28] Irene Di Ceglie;Silvia Carnevale;Anna Rigatelli;Giovanna Grieco;Piera Molisso;Sebastien Jaillon. Immune cell networking in solid tumors: focus on macrophages and neutrophils.. Frontiers in immunology(IF=5.9). 2024. PMID:38426089. DOI: 10.3389/fimmu.2024.1341390.
- [29] Fujin Fang;Tiantian Zhang;Qiong Li;Xiaowei Chen;Fei Jiang;Xiaobing Shen. The tumor immune-microenvironment in gastric cancer.. Tumori(IF=3.1). 2022. PMID:35196917. DOI: 10.1177/03008916211070051.
- [30] Katrin Pansy;Barbara Uhl;Jelena Krstic;Marta Szmyra;Karoline Fechter;Ana Santiso;Lea Thüminger;Hildegard Greinix;Julia Kargl;Katharina Prochazka;Julia Feichtinger;Alexander Ja Deutsch. Immune Regulatory Processes of the Tumor Microenvironment under Malignant Conditions.. International journal of molecular sciences(IF=4.9). 2021. PMID:34948104. DOI: 10.3390/ijms222413311.
- [31] Ye Lu;Jiarong Zheng;Pei Lin;Yunfan Lin;Yucheng Zheng;Zizhao Mai;Xu Chen;Tian Xia;Xinyuan Zhao;Li Cui. Tumor Microenvironment-Derived Exosomes: A Double-Edged Sword for Advanced T Cell-Based Immunotherapy.. ACS nano(IF=16.0). 2024. PMID:39319751. DOI: 10.1021/acsnano.4c09190.
- [32] Jingyao Hu;Xinyu Li;Liang Yang;Hangyu Li. Hypoxia, a key factor in the immune microenvironment.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2022. PMID:35676780. DOI: 10.1016/j.biopha.2022.113068.
- [33] Zhipeng Li;Fang Ning;Changduo Wang;Hongli Yu;Qingming Ma;Yong Sun. Normalization of the tumor microvasculature based on targeting and modulation of the tumor microenvironment.. Nanoscale(IF=5.1). 2021. PMID:34651623. DOI: 10.1039/d1nr03387e.
- [34] Pablo Igor Ribeiro Franco;Arthur Perillo Rodrigues;Liliana Borges de Menezes;Marina Pacheco Miguel. Tumor microenvironment components: Allies of cancer progression.. Pathology, research and practice(IF=3.2). 2020. PMID:31735322. DOI: 10.1016/j.prp.2019.152729.
- [35] Xianjie Jiang;Jie Wang;Xiangying Deng;Fang Xiong;Shanshan Zhang;Zhaojian Gong;Xiayu Li;Ke Cao;Hao Deng;Yi He;Qianjin Liao;Bo Xiang;Ming Zhou;Can Guo;Zhaoyang Zeng;Guiyuan Li;Xiaoling Li;Wei Xiong. The role of microenvironment in tumor angiogenesis.. Journal of experimental & clinical cancer research : CR(IF=12.8). 2020. PMID:32993787. DOI: 10.1186/s13046-020-01709-5.
- [36] Geetika Kaur;Bipradas Roy. Decoding Tumor Angiogenesis for Therapeutic Advancements: Mechanistic Insights.. Biomedicines(IF=3.9). 2024. PMID:38672182. DOI: 10.3390/biomedicines12040827.
- [37] Dasom Kim;Kyeong Seob Hwang;Eun U Seo;Suyeong Seo;Byung Chul Lee;Nakwon Choi;Jonghoon Choi;Hong Nam Kim. Vascularized Lung Cancer Model for Evaluating the Promoted Transport of Anticancer Drugs and Immune Cells in an Engineered Tumor Microenvironment.. Advanced healthcare materials(IF=9.6). 2022. PMID:35286780. DOI: 10.1002/adhm.202102581.
- [38] Nicole M Anderson;M Celeste Simon. The tumor microenvironment.. Current biology : CB(IF=7.5). 2020. PMID:32810447. DOI: 10.1016/j.cub.2020.06.081.
- [39] Dhruba Kadel;Yu Zhang;Hao-Ran Sun;Yue Zhao;Qiong-Zhu Dong;Lun-Xiu Qin. Current perspectives of cancer-associated fibroblast in therapeutic resistance: potential mechanism and future strategy.. Cell biology and toxicology(IF=5.9). 2019. PMID:30680600. DOI: 10.1007/s10565-019-09461-z.
- [40] Kim H T Paraiso;Keiran S M Smalley. Fibroblast-mediated drug resistance in cancer.. Biochemical pharmacology(IF=5.6). 2013. PMID:23376122. DOI: .
- [41] Li V Yang. Tumor Microenvironment and Metabolism.. International journal of molecular sciences(IF=4.9). 2017. PMID:29258182. DOI: 10.3390/ijms18122729.
- [42] María Belén Novoa Díaz;María Julia Martín;Claudia Gentili. Tumor microenvironment involvement in colorectal cancer progression via Wnt/β-catenin pathway: Providing understanding of the complex mechanisms of chemoresistance.. World journal of gastroenterology(IF=5.4). 2022. PMID:36051330. DOI: 10.3748/wjg.v28.i26.3027.
- [43] Naji Kharouf;Thomas W Flanagan;Sofie-Yasmin Hassan;Hosam Shalaby;Marla Khabaz;Sarah-Lilly Hassan;Mosaad Megahed;Youssef Haikel;Simeon Santourlidis;Mohamed Hassan. Tumor Microenvironment as a Therapeutic Target in Melanoma Treatment.. Cancers(IF=4.4). 2023. PMID:37370757. DOI: 10.3390/cancers15123147.
- [44] Oliwia Piwocka;Igor Piotrowski;Wiktoria M Suchorska;Katarzyna Kulcenty. Dynamic interactions in the tumor niche: how the cross-talk between CAFs and the tumor microenvironment impacts resistance to therapy.. Frontiers in molecular biosciences(IF=4.0). 2024. PMID:38455762. DOI: 10.3389/fmolb.2024.1343523.
- [45] Giuseppina Roscigno;Sacha Jacobs;Belén Toledo;Roberto Borea;Gianluca Russo;Francesco Pepe;Maria Jose Serrano;Viola Calabrò;Giancarlo Troncone;Roberto Giovannoni;Elisa Giovannetti;Umberto Malapelle. The potential application of stroma modulation in targeting tumor cells: Focus on pancreatic cancer and breast cancer models.. Seminars in cancer biology(IF=15.7). 2025. PMID:40373890. DOI: 10.1016/j.semcancer.2025.05.003.
- [46] Lu Liu;Yafei Li;Bo Li. Interactions between cancer cells and tumor-associated macrophages in tumor microenvironment.. Biochimica et biophysica acta. Reviews on cancer(IF=8.3). 2025. PMID:40345263. DOI: 10.1016/j.bbcan.2025.189344.
- [47] Sri Sathya Sandilya Garemilla;Siri Chandana Gampa;Sireesha Garimella. Role of the tumor microenvironment in cancer therapy: unveiling new targets to overcome drug resistance.. Medical oncology (Northwood, London, England)(IF=3.5). 2025. PMID:40332723. DOI: 10.1007/s12032-025-02754-w.
- [48] Mohamed El-Tanani;Syed Arman Rabbani;Rasha Babiker;Imran Rangraze;Sumedha Kapre;Sushesh Srivastsa Palakurthi;Abdullah M Alnuqaydan;Alaa A Aljabali;Manfredi Rizzo;Yahia El-Tanani;Murtaza M Tambuwala. Unraveling the tumor microenvironment: Insights into cancer metastasis and therapeutic strategies.. Cancer letters(IF=10.1). 2024. PMID:38626856. DOI: 10.1016/j.canlet.2024.216894.
- [49] Pratikshya Sa;Sanjeeb K Sahoo;Fahima Dilnawaz. Responsive Role of Nanomedicine in the Tumor Microenvironment and Cancer Drug Resistance.. Current medicinal chemistry(IF=3.5). 2023. PMID:36154585. DOI: 10.2174/0929867329666220922111336.
MaltSci Intelligent Research Services
© 2025 MaltSci
