Skip to main content

Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota

Abstract

Background

Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment.

Methods

Keywords such as “prostate cancer”, “tumor microenvironment”, “immune cells”, “vascular system”, “stromal cells”, and “microbiota” were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment.

Results

The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment.

Conclusions

A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.

Graphical Abstract

Introduction

Prostate cancer (PCa) is among the most common cancers in men globally. Global cancer statistics report approximately 1.4 million new cases and 375,000 deaths annually. The incidence of PCa rises significantly in men over 65, severely impacting their health and quality of life [1]. Recent research from China and the United States indicates a rapid increase in PCa incidence among men aged 25–34. Younger men diagnosed with PCa are more likely to develop aggressive forms of the disease, leading to lower five-year survival rates [2]. Metastatic PCa, compared to non-metastatic PCa, is associated with significantly higher morbidity and mortality. Furthermore, many patients with non-metastatic PCa eventually progress to castration-resistant prostate cancer (CRPC) after undergoing androgen deprivation therapy (ADT). CRPC presents greater treatment challenges and is linked to a poorer prognosis and reduced survival outcomes [3]. Over the past few decades, clinical research on PCa has been a focal point in the medical field [4]. The emergence of the Human Genome Project alongside high-throughput technologies has significantly deepened our grasp of PCa’s genetic underpinnings, marking a pivotal focus in research endeavors [5, 6]. However, focusing solely on the genetic level is insufficient to understand the complexity of PCa. There is increasing recognition that studying the PCa microenvironment is crucial for PCa research, and its comprehensive impact is becoming more prominent [7, 8]. PCa is not only caused by genetic mutations but also by interactions with its surrounding microenvironment. The PCa microenvironment comprises multiple components, including immune cells, vascular systems, stromal cells, and microorganisms, which interact to form a highly dynamic and interconnected ecosystem [9,10,11,12]. A deep understanding of the structure and function of this complex network is crucial for elucidating the pathogenesis of PCa. In PCa, the tumor microenvironment (TME) exhibits astonishing levels of hierarchy and diversity [13]. The immune cells exhibit diverse types and functions, with specific T lymphocyte subsets essential for immune surveillance and anti-tumor responses [14]. The vascular endothelial growth factor (VEGF) family, part of the angiogenesis pathway, closely associates with the tumor’s blood supply and nutrient delivery, directly impacting tumor growth and spread [15]. Stromal cells exert an impact on tumor invasion and metastasis through modulation of the extracellular matrix (ECM) [16]. Moreover, the connection between the microbiota and PCa is still developing [17]. The adoption of emerging technologies, such as single-cell sequencing, offers significant potential to uncover the intricate details of the PCa microenvironment [18]. This is essential for gaining a comprehensive understanding of PCa and establishing a robust foundation for the development of future therapeutic strategies (Fig. 1).

Fig. 1
figure 1

TME interaction: an ecological perspective on PCa. Single-cell sequencing technology provides a thorough perspective of the PCa microenvironment. The stromal cells, comprising endothelial cells, epithelial cells, pericytes, and CAFs, undergo varying degrees of alterations within the microenvironment of PCa. The M2-macrophages (M2-MФ) subset of macrophages exhibits significant dynamic changes, underscoring the crucial role of immune regulation in this context. Tumor-inflammatory monocytes (TIMo), as one of the monocyte subpopulations, also demonstrates high levels of variation, further highlighting the immune system’s response to PCa. B cells are activated, potentially participating in immune response regulation. However, NK cells activity exhibits a decreasing trend, whereas CTL cells demonstrate significant activity changes, indicating potential distinct roles and regulation of immune cells in responding to PCa

The clinical significance of the microenvironment in diagnosing, treating, and predicting the prognosis of PCa is becoming more prominent [19]. The potential use of microenvironmental biomarkers has become crucial for early diagnosis and treatment monitoring [20]. The tumor immune response is closely linked to patients’ survival rates, making immunotherapy a promising approach for PCa treatment [21]. Additionally, targeted drug research on the microenvironment is ongoing, offering more opportunities for precise treatment [22]. Recent advances in PCa treatment, especially those targeting the TME, have introduced new possibilities for precision therapy [23]. Drugs targeting VEGF inhibit angiogenesis in PCa, reshaping the TME to limit cancer growth and metastasis [24]. Programmed cell death-ligand 1 (PD-L1) inhibitors, such as atezolizumab, enhance the immune system’s attack on tumors, providing long-term survival benefits for some PCa patients, especially those with treatment-resistant cases [25, 26]. Traditional non-specific chemotherapeutic agents, like cisplatin and 5-fluorouracil, have limited effects on non-essential molecules within the PCa microenvironment and exhibit significant toxicity, leading to suboptimal anti-tumor effects in clinical practice [27, 28]. While these agents can temporarily suppress tumor growth, prolonged use may lead to adaptive changes in tumors, causing resistance, reduced treatment efficacy, or recurrence. Therefore, when formulating individualized treatment plans, it is crucial to consider the PCa microenvironment and the patient’s pathological features. Target key factors involved in tumor growth and metastasis while avoiding excessive intervention on non-essential microenvironment components. This approach aims to maximize therapeutic efficacy, minimize unnecessary side effects, and improve patient survival and quality of life. Another significant challenge in precision therapy for PCa is the variability in individual treatment responses. Different patients exhibit varying responses to the same therapeutic regimen, emphasizing the need for personalized treatment strategies. Integrating multi-omics data, including genomics, transcriptomics, and proteomics, enhances our understanding of the molecular mechanisms underlying PCa, leading to more targeted and effective treatments [29]. Looking ahead, research on the PCa microenvironment still encounters numerous challenges. Continuous innovation in technical methods and research approaches will delve deeper into the mysteries of the microenvironment. Overall, this review will explore various aspects of the PCa microenvironment, aiming to provide readers with a comprehensive and clear overview. By gaining a comprehensive understanding of the complexity of the PCa microenvironment, new insights and inspirations are anticipated to enhance future personalized treatment and clinical interventions.

PCa microenvironment characteristics

Relationship between immune cells and the PCa microenvironment

Immune cells in the PCa microenvironment, including T and B lymphocytes, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), play a critical role [29, 30]. The role of the androgen receptors (AR) in the PCa immune microenvironment is significant. AR signaling regulates immune cells, particularly T cells and natural killer (NK) cells, and has a profound impact on tumor immune surveillance. Studies have shown that AR signaling upregulates PD-L1 expression, weakening the anti-tumor functions of T cells and NK cells, thereby inhibiting the body’s immune response against the tumor [31, 32]. Additionally, AR modulates T cell exhaustion, further enhancing the tumor’s ability to evade immune surveillance. AR not only directly affects immune cells but also modifies immune-regulatory factors within the TME, promoting the formation of an immunosuppressive environment. Therefore, AR acts as a critical regulator of immune evasion, playing a pivotal role in the progression of PCa.

MDSCs are aberrantly activated, immature, and heterogeneous immunosuppressive cells significantly enriched in the PCa microenvironment, impacting PCa development and metastasis [33]. Preclinical in vivo study shows that loss of ARID1A function in mouse models induces MDSCs recruitment by activating the nuclear factor-kappa B (NF-κB) signaling pathway, reduces CD8+ T cells infiltration, creates an immunosuppressive TME, and promotes PCa progression [33]. Clinical studies indicate that MDSCs numbers in the peripheral blood of PCa patients are significantly higher than in healthy donors and negatively correlate with patient survival. Interleukin-23 (IL-23) secreted by MDSCs plays a pivotal role in driving the progression and metastasis of CRPC. Mechanistically, IL-23 inhibition suppresses AR signaling, which in turn reduces the survival and proliferation of PCa cells. Additionally, targeting IL-23 can restore sensitivity to ADT in patients with advanced PCa. Consequently, immunotherapies that block MDSCs recruitment or directly inhibit IL-23 offer promising therapeutic strategies for treating these aggressive and prevalent malignancies [34]. The micro-scale interactions between T lymphocytes and antigen-presenting cells (APCs) in the PCa microenvironment are crucial for understanding PCa development and the anti-cancer immune response [35, 36]. Complex regulatory mechanisms widely recognized influence the differentiation and activation status of T lymphocytes [37, 38]. The importance of APCs in the immune surveillance of PCa has been extensively studied, revealing their key role in tumor immune escape through intricate interaction networks [36]. These interactions involve signaling networks between APCs and T lymphocytes, as well as interactions with PCa cells. These novel findings deepen our understanding of APCs’ functions and provide important clues for designing precise immune therapies targeting the PCa microenvironment. These findings lay a scientific groundwork for the development of more potent cancer treatment strategies and pave the way for further advancements in cancer immunotherapy (Fig. 2).

Fig. 2
figure 2

Immune cells’ dynamic interplay in PCa microenvironment. DCs are the most powerful professional APCs with immunostimulatory properties. Despite impaired cellular phagocytic functions, tumor antigens can still be presented to T cells by DC cells. This process involves handling and presenting tumor antigens to T cells, facilitating T cells recognition. CD4+ T cells and CD8+ T cells are then activated and mobilized to infiltrate the PCa site. During this period, PCa patients exhibit an increase in mature and activated B cells within the tumor-draining SNs. Additionally, the immune response in PCa patients who receives ADT is enhanced by promoting the recruitment of TAMs and activating CD8+ T cells. Upon recognizing cancer cells, T cells initiate cytotoxicity against them, promoting the immune system’s antitumor response to PCa

Subgroups and functions of T lymphocytes

PCa tissues contain various T lymphocyte subgroups, each with distinct roles and functions in the PCa microenvironment. The study of helper CD4+ T (Th) cells, regulatory CD4+ T (Treg) cells, and cytotoxic CD8+ T lymphocyte (CTL) cells has received significant attention [39,40,41]. Th1 cells, pro-inflammatory CD4+ T cells, support CD8+ T cells by secreting interleukin-2 (IL-2) and interferon-γ (IFN-γ). An increase in Th1 cells in the PCa microenvironment is closely associated with a favorable prognosis. In contrast, Th2 cells can drive tumor progression, therapeutic resistance, and castration resistance by producing transforming growth factor-β (TGF-β) or prostaglandin E2 (PGE2), leading to poor outcomes for PCa patients [42]. Treg cells essential for suppressing inflammatory responses and controlling autoimmunity, are ubiquitous in the PCa microenvironment. They promote tumor development and progression by inhibiting anti-tumor immune responses. For example, Treg cells secrete IL-2 to regulate NK cell homeostasis and function. Treg cells also support PCa cells survival directly through growth factor secretion and indirectly by interacting with stromal cells, such as cancer-associated fibroblasts (CAFs). Thus, infiltrating Treg cells can serve as significant adverse prognostic markers for PCa recurrence [43, 44]. CTL cells recognize abnormal tumor antigens on PCa cells and target them for destruction. After killing PCa cells, CTL cells also inhibit angiogenesis by secreting IFN-γ [45,46,47]. However, other CD8+ T cells may be inhibited, possibly due to the influence of inhibitory cells and molecules in the microenvironment. This inhibition may lead to functional exhaustion of CD8+ T cells, limiting their anti-tumor effects. Additionally, complex interactions occur among T cells subgroups within the microenvironment [48]. The synergistic effects between CD4+ T cells and CD8+ T cells may critically regulate immune responses. However, the molecular mechanisms of this synergy are not fully understood and require further investigation [49]. The diversity and complexity of T lymphocyte subgroups within the PCa microenvironment necessitate a deeper comprehension of their specific functions, which will enhance the development of more effective immunotherapy strategies for PCa.

APCs

APCs, including dendritic cells (DCs), TAMs, and B cells, are crucial in the immunoregulation of PCa [50, 51]. The immune system’s main function is to identify and eliminate abnormal cells, including cancer cells [52]. In this process, APCs present antigens and activate immune cells, but must first be activated. Once activated, DCs absorb, process, and present antigens from PCa cells. These DCs relay this information to nearby lymph nodes, activating naive T cells [53]. Activated T cells then identify and attack PCa cells, resulting in an immune cytotoxic effect [54, 55]. DCs are the most potent professional APCs with immunostimulatory capabilities, and the only APCs capable of activating naive T cells. However, research has demonstrated that PCa cells promote the recruitment of immature DCs to tumor sites by inducing inflammatory chemokine production. This recruitment leads to molecular and signaling alterations in DCs, impairing their antigen-presenting function and ultimately suppressing antitumor immune responses [56]. As the immune response progresses, activated TAMs play a crucial role. They present antigens to engaged T cells, promoting a sustained “high-energy state” through repeated stimulation, enhancing T cells’ ability to combat PCa. A Study has shown that after receiving ADT, immune responses are enhanced in PCa patients by recruiting TAMs and activating CD8+ T cells, leading to the death of PCa cells [57]. Additionally, APCs contribute to immune memory formation. Upon activation against PCa, the immune system retains a specific pool of T and B cells. In subsequent encounters with the same antigen, memory B cells rapidly gather small amounts of antigen to activate T cells, resulting in a faster, more effective response [58]. In PCa patients, mature and activated B cells are elevated in the tumor-draining sentinel lymph nodes (SNs), promoting B cell-specific antitumor immune responses [59]. APCs modulate the immune response’s magnitude and orientation through the secretion of cytokines like interleukins (IL) and tumor necrosis factor (TNF). This process aids in maintaining immune system equilibrium, ensuring an effective response against PCa cells while preventing excessive immune reactions [60,61,62].

Relationship between angiogenesis and PCa

Angiogenesis and PCa interaction are heavily researched topics. Recent studies show a strong link between angiogenesis and PCa [63, 64]. The increase in newly formed blood vessels in PCa tissues is closely linked to PCa growth and invasion. These vessels supply nutrients and oxygen to cancer cells and help evade immune surveillance, promoting continuous PCa growth [65]. Additionally, the network of new vessels aids cancer cells metastasis, leading to distant metastatic lesions and affecting PCa prognosis. PCa cells activate multiple angiogenesis-related signaling pathways, such as VEGF, fibroblast growth factor (FGF), and platelet-derived growth factor (PDGF) [66,67,68,69]. Growth factors and their corresponding receptors are significantly expressed in PCa tissues, promoting new blood vessel formation through the regulation of endothelial cells activities, including proliferation, migration, and lumen formation, among other biological processes. Moreover, studies indicate that oncogene activation and tumor suppressor gene inactivation also contribute to angiogenesis regulation, emphasizing the strong link between angiogenesis and PCa [70]. Extensive research has also uncovered the significant contribution of cytokines and ECM in the PCa microenvironment during angiogenesis. PCa cells interact with surrounding stromal cells, releasing various cytokines such as interleukin-6 (IL-6), and interleukin-30 (IL-30). These cytokines not only directly promote angiogenesis but also indirectly regulate new blood vessel formation by activating inflammatory responses and altering stromal properties, among other pathways [71, 72]. Moreover, tumor-related angiogenesis is influenced by microenvironmental factors like hypoxia and acidic environments. These factors further increase PCa cells’ reliance on angiogenesis [73,74,75]. A deeper understanding of the relationship between angiogenesis and PCa aids in clarifying PCa’s pathogenesis and provides a theoretical foundation for developing targeted anti-angiogenesis therapies. Further research in this field is anticipated to offer new insights and directions for future clinical treatments, potentially enhancing therapeutic options for patients (Fig. 3).

Fig. 3
figure 3

Vascular networks in PCa: exploring angiogenesis’s impact. The VEGF family comprises various subtypes, including VEGF-A, VEGF-B, VEGF-C, VEGF-D, and PLGF. VEGF-A binds to TYR kinase receptors, activating signaling pathways such as Ras/MAPK and PI3K/Akt, thereby promoting angiogenesis. VEGF regulates the activity of TAMs and T lymphocytes, impacting the distribution and function of immune cells within the PCa microenvironment and influencing the immune escape mechanisms of PCa cells. VEGF supplies nutrients and oxygen essential for cancer cells invasion and metastasis, influencing critical processes such as cells adhesion and basement membrane degradation, thereby facilitating the migration and spread of PCa cells. The FGF family mediates cells communication through cells surface receptors, promoting cancer cells proliferation and survival, regulating the expression of cells cycle proteins, and influencing cells mitosis and growth. FGF also contributes to angiogenesis, supplying oxygen and nutrients to cancer cells, thus promoting cancer cells growth and spread. Members of the PDGF family drive the spread of PCa cells to surrounding tissues by regulating interactions between cells and degrading the ECM. PCa cells often enhance angiogenesis by producing PDGF, which increases VEGF expression and indirectly regulates vascular development

In addition, the AR plays a crucial role in the progression of PCa by regulating angiogenesis. Angiogenesis is a key process by which tumors acquire oxygen and nutrients to promote growth and metastasis, with VEGF serving as a central regulatory factor. Studies have shown that AR signaling can upregulate the expression of VEGF in the TME, thereby promoting the proliferation and migration of endothelial cells and enhancing the formation of new blood vessels [76, 77]. Moreover, AR-mediated signaling pathways not only regulate the speed and quality of tumor blood vessel formation but also significantly increase the tumor’s blood supply, further driving rapid tumor progression. Specifically, in PCa, AR regulates the expression of VEGF and other angiogenesis-related genes to maintain the vascular network necessary for tumor growth [32]. This mechanism indicates that AR not only controls the proliferation of PCa cells but also accelerates the deterioration of the TME by influencing angiogenesis.

The role of the VEGF family

The VEGF family is identified as the most potent and crucial among the numerous factors involved in angiogenesis. Multiple factors in the PCa microenvironment can trigger VEGF production and subsequent angiogenesis. The VEGF family includes subtypes like vascular endothelial growth factor-A (VEGF-A), vascular endothelial growth factor-B (VEGF-B), vascular endothelial growth factor-C (VEGF-C), vascular endothelial growth factor-D (VEGF-D), and placental growth factor (PLGF), with varying expression and functions in PCa. This diversity offers extensive evidence for investigating their mechanisms of action [78, 79]. Among these, VEGF-A is extensively studied and binds to the tyrosinase (TYR) kinase receptor, activating signaling pathways like ras-mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), promoting angiogenesis [80]. However, the functions of VEGF and its subtypes like PLGF in the PCa microenvironment are not fully understood, although study suggests their potential roles in regulating tumor angiogenesis, metastasis, and drug resistance [81]. VEGF regulates TAMs and T lymphocytes, shaping immune cell distribution and function within the TME. This modulation directly influences immune cell behavior and the tumor’s immune evasion strategies, ultimately impacting overall immune responses [82]. Additionally, the VEGF family is closely associated with tumor cells invasion and metastasis in the PCa microenvironment [83]. VEGF promotes angiogenesis, supplying essential nutrients and oxygen for cancer cells invasion and metastasis. Its influence on critical processes like cells adhesion and basement membrane degradation facilitates favorable conditions for PCa cells migration and spread. Research in this field enhances understanding of PCa development mechanisms, providing new targets and treatment strategies to inhibit tumor invasion and metastasis [67, 81, 84]. Inhibitors targeting the VEGF family are now first-line drugs for PCa treatment [15, 85]. Challenges such as PCa cells resistance and treatment side effects continue. Thus, a thorough understanding of the detailed mechanisms of the VEGF family in the PCa microenvironment can precisely regulate its function, improve treatment effectiveness, and establish more reliable foundations for personalized PCa patient treatment. The significant involvement of the VEGF family in the PCa microenvironment encompasses various aspects like signaling pathways, immune regulation, invasion, and metastasis, offering extensive research evidence for a better grasp of PCa development mechanisms and the development of more effective treatment strategies.

Roles of FGF family

FGF, unlike VEGF, acts on various cell types, directly or indirectly stimulating angiogenesis as a mitogen specific to endothelial cells. The FGF family comprises proteins involved in embryonic development, angiogenesis, wound healing, and cancer progression, exhibiting paracrine, autocrine, or endocrine functions [86]. The FGF family, crucial in regulating signaling pathways, has distinct functions in PCa progression [87, 88]. Binding to PCa cells surface receptors, the FGF family facilitates intercellular communication, promoting PCa cells growth and survival [89]. FGF activation specifically regulates cells cycle protein expression, impacting cells division and growth. This precise regulation is crucial for PCa initiation and progression [90]. Most studies have focused on FGF1 and FGF2 among the several types comprising the FGF family. FGF1 can induce matrix metalloproteinases (MMPs) expression in PCa cells, contributing to malignant progression. FGF2 directly stimulates angiogenesis and plays a role in cancer by inducing cell migration, proliferation, and differentiation [91, 92]. In addition, the FGF family inhibits the transition of PCa to CRPC. Studies have shown that SOX2 is considered to be a regulator of FGF expression, and in CRPC cells, ectopic expression of SOX2 leads to an 18.81-fold increase in FGF levels [93, 94]. This immune evasion promotes PCa development, potentially causing resistance and recurrence. Overall, the diverse roles of the FGF family in the PCa microenvironment offer a new perspective on cancer initiation and progression. Accurately understanding its mechanisms in cells proliferation, angiogenesis, and immune regulation aids in precise PCa interventions and treatments, offering strong theoretical support for future therapies.

The role of the PDGF family

PDGF, a peptide regulatory factor primarily secreted by macrophages, stimulates tissue cell growth. Its role in angiogenesis is weaker than that of FGF and VEGF, suggesting it may not be essential for the initial formation of blood vessels. Under physiological conditions, PDGF resides in platelets and is released upon platelet activation during blood coagulation, stimulating specific cell chemotaxis and growth. The PDGF family regulates tumor growth and metastasis through interactions with PCa cells. Specifically, PDGF family members activate multiple signaling pathways by binding to receptors on cancer cells surfaces, leading to cancer cells proliferation [95, 96]. A detailed understanding of this mechanism provides new insights into unraveling the molecular regulatory network of PCa cells growth. Additionally, the PDGF family is crucial for PCa invasion and metastasis. PDGF family members promote the spread of PCa cells to surrounding tissues by regulating cell-cell interactions and matrix degradation processes [97, 98]. This discovery deepens our understanding of PCa metastasis mechanisms and provides new targets for intervention in this process. The regulation of angiogenesis in the PCa microenvironment by the PDGF family is of great interest. Tumor cells typically promote angiogenesis by releasing PDGF, which upregulates VEGF expression and indirectly mediates vascular formation. The complex regulatory network of neovascularization provides profound insights for designing more precise anti-angiogenic therapy strategies [99]. In summary, the PDGF family’s role in the PCa microenvironment encompasses cells proliferation, invasion, and angiogenesis. A detailed understanding of these mechanisms provides a theoretical basis for future treatment strategies. These research findings provide new perspectives for deepening our understanding of the biological characteristics of PCa and offer strong support for targeted therapy in clinical applications.

Interaction between endothelial cells and PCa cells

Endothelial cells directly influence PCa cells proliferation and invasion by secreting growth factors like VEGF and MMPs [100, 101]. These factors stimulate tumor cells growth and regulate TME acidity, oxygen levels, and consequently, metabolic adaptation and drug sensitivity. Additionally, interactions between endothelial cell surface receptors and those on PCa cells significantly influence tumor biology [102]. Integrin binding between endothelial and PCa cells fosters PCa cells reliance on the microenvironment and activates intracellular signaling pathways, thereby affecting cells migration and infiltration capabilities [103]. Endothelial cells in the PCa microenvironment regulate PCa cells behavior through exosome release [102, 104, 105]. Exosomes carry miRNAs, proteins, and biomolecules that impact gene expression and cells signaling in PCa cells through transport. This offers new insights into the complex interactions between PCa and endothelial cells.

Functions of stromal cells and their impacts on PCa development

Stromal cells are essential for maintaining the structure and function of prostate tissue [106]. They maintain ECM integrity by synthesizing matrix molecules like collagen and elastic fibers, creating a supportive environment for normal prostate cells [107, 108]. However, during PCa development, this supportive function also lays the stromal groundwork for the infiltration and spread of PCa cells, facilitating their invasion into surrounding tissues. In addition to providing structural support, stromal cells also modulate inflammatory responses and immune reactions. The inflammatory state in PCa is considered a key factor in cancer initiation and progression. Stromal cells directly or indirectly influence immune regulation within the PCa microenvironment through the production of inflammatory cytokines, chemokines, and modulation of immune cells activity. These immune regulatory changes may suppress PCa immune responses, contributing to immune evasion in PCa [109, 110]. Stromal cells also contribute to angiogenesis by secreting angiogenesis-related factors such as VEGF, promoting the formation of new blood vessels that supply tumors with oxygen and nutrients. This process is crucial for the growth and metastasis of PCa and provides a potential target for anti-angiogenic therapies.

In this context, CAFs, as one of the most important stromal cells in the TME, play a particularly prominent role in PCa. CAFs promote PCa cell proliferation, invasion, and migration by secreting growth factors, cytokines, and matrix remodeling proteins. AR plays a key role in regulating the function of CAFs. AR signaling directly or indirectly modulates the gene expression of CAFs, altering the secretion of bioactive substances, and thereby promoting tumor progression [111]. For example, AR can upregulate the expression of cytokines such as TGF-β, inducing CAFs to secrete more ECM, which enhances tumor cell invasiveness and drug resistance [112]. Furthermore, CAFs can regulate other components of the TME, such as angiogenesis and immune cell function, through AR signaling, further exacerbating the malignant behavior of the tumor. Therefore, CAFs, as key regulators of the tumor stroma, provide multiple layers of support for PCa development through their interaction with AR signaling.

The presence of stromal cells also significantly impacts the therapeutic response in PCa. Due to their involvement in forming and maintaining the PCa microenvironment, stromal cells may affect the delivery and efficacy of anticancer drugs [22]. Additionally, stromal cells may directly or indirectly influence treatment outcomes by regulating drug sensitivity and resistance in PCa. These findings underscore the importance of considering the multifaceted roles and impacts of stromal cells in the PCa microenvironment when developing treatment strategies for PCa (Fig. 4).

Fig. 4
figure 4

Decoding stromal cells: key players in PCa evolution. CAFs and adipocytes produce various ECM components and proteases such as MMPs. Some ECM components degrade, while others harden over time through continuous remodeling. PCa cells can activate CAFs by secreting IL-6, thereby promoting EMT and angiogenesis of PCa cells; Adipocytes can transform into malignant adipocytes, providing enhanced support and supply to PCa cells. Hypoxia-inducible factor stimulates endothelial cells to secrete pro-angiogenic factors like PDGF and VEGF, initiating the process of angiogenesis

Roles of CAFs and adipocytes

Stromal cells, such as CAFs and adipocytes, are crucial in the PCa microenvironment [113, 114]. CAFs, located primarily in stromal tissue, maintain tissue structure, regulate cell signaling, and participate in cell proliferation and differentiation. Within the PCa microenvironment, CAFs and PCa cells interact, with PCa cells activating CAFs by secreting IL-6. Once activated, CAFs promote the epithelial-mesenchymal transition (EMT) of PCa cells and secrete growth factors that enhance angiogenesis, supplying PCa cells with nutrients and oxygen, thus fostering their growth [115,116,117]. Recent studies indicate that CAFs exhibit higher levels of autophagy than normal fibroblasts (NFs) and promote the malignant phenotype of PCa through autophagy-related gene 5 (ATG5) dependent autophagy [76, 118]. CAFs also influence the immune system’s surveillance and clearance of cancer cells by modulating the activity and distribution of immune cells. These changes in immune regulation help cancer cells evade immune surveillance within the host, enhancing their survival and dissemination opportunities. Additionally, adipocytes, another type of stromal cells, contribute significantly to the PCa microenvironment [119]. Adipocytes, primarily located in adipose tissue, are responsible for storing and releasing fat and regulating energy metabolism [120]. During the progression of PCa, adipocytes can transform within the PCa microenvironment to become malignant adipocytes. This transformation is influenced by various factors secreted by PCa cells and CAFs, which induce changes in the function and phenotype of adipocytes. For instance, the release of inflammatory cytokines and alterations in signaling pathways can lead to the recruitment and activation of adipocyte precursor cells, which then differentiate into malignant adipocytes [121]. Malignant adipocytes release significant amounts of lipid metabolites and fatty acids, providing essential energy and nutrients for PCa cells growth and metastasis [122]. Thus, CAFs and adipocytes in the PCa microenvironment play roles beyond PCa support, involving various mechanisms in tumor growth, dissemination, and immune evasion.

The role of ECM

The ECM’s role in the PCa microenvironment has attracted significant attention and is a recent focus of research [123]. The ECM mainly consists of collagen, fibronectin, elastin, and glycoproteins like laminin. Interactions between cancer cells and stromal cells alter ECM components, affecting cancer cell invasion and metastasis [124]. Cancer cells recruit stromal cells like CAFs and adipocytes, which collaboratively remodel the ECM. These stromal cells produce and release various ECM components and proteases, including MMPs. Some ECM components degrade, while others harden over time through continuous remodeling. Increasing evidence shows that ECM stiffening drives PCa growth, invasion, and metastasis. Clinically, malignant lesions are often stiffer than benign tissues. This finding offers novel biomarkers for predicting PCa progression and metastasis. Additionally, ECM stiffening may crucially affect the PCa microenvironment by modulating signaling pathways. Numerous studies have shown that key pathways in CRPC, such as PI3K/Akt and MAPK, are activated by ECM stiffening [125, 126]. The ECM’s role in the PCa microenvironment goes beyond traditional support and structure, uncovering complex and diverse functions. These studies enhance our comprehension of ECM’s role in the PCa microenvironment and provide general insights for understanding and intervening in PCa microenvironments.

The relationship between microorganisms and PCa

In previous research, microbiologists and cancer researchers have increasingly turned their attention to the biomedical aspects of the relationship between bacteria, viruses, and PCa [17, 127]. Recently, advanced molecular biology and high-throughput sequencing technologies have allowed us to recognize the potentially crucial regulatory role of the microbiota in the etiology and progression of PCa [128]. The relationship between PCa and bacteria has garnered significant attention, particularly in earlier studies that emphasized the potential role of urethral bacteria in PCa development [129]. Nevertheless, recent studies have identified a specific microbial community in prostate tissue, comprising distinct bacterial subgroups [130, 131]. These bacteria may significantly impact PCa development by activating inflammatory pathways, influencing immune responses, or directly interfering with the cells cycle [132]. This finding emphasizes the intricate relationship between bacteria and PCa, offering a fresh perspective for investigating the specific role of prostate microbiota. Simultaneously, viruses are also implicated in the pathogenic mechanisms of PCa [133]. Virus infections have been linked to various cancer developments, with specific interest among researchers in certain viruses found in PCa, such as human papilloma virus (HPV), epstein-barr virus (EBV), and the human herpes virus (HHV) [134,135,136]. These viruses may contribute to PCa pathogenesis through multiple pathways, such as direct interference with cells genome stability, impact on cells survival signaling pathways, and interaction with the host immune system. Thus, the presence of viruses can significantly impact the pathophysiological processes of PCa. It is important to note that the relationship between microorganisms and PCa remains complex and challenging to research [129, 137]. Future investigations should employ extensive experimental designs and conduct large-scale human studies to substantiate these connections, thereby enhancing our insights into the involvement of microorganisms in the onset and progression of PCa. This not only uncovers potential pathogenic mechanisms of PCa but also establishes a solid foundation for developing new prevention and treatment strategies (Fig. 5).

Fig. 5
figure 5

Exploring the intricate connection between microorganisms and PCa. Upon viral infection, normal cells may undergo integration of the virus into their DNA, resulting in apoptosis or transformation into pre-cancerous cells. Bacterial stimulation elicits inflammatory responses that can activate IL-15, influencing neutrophil activity and impacting PCa progression

Despite the increasing focus on the relationship between microorganisms and PCa, the connection between AR and gut microbiota has not been extensively studied. However, recent evidence suggests that AR may indirectly regulate the composition of gut microbiota by influencing the host’s immune and metabolic states, thus impacting the development of PCa. Studies indicate that androgen signaling could alter the body’s metabolic activities and oxidative defense systems, thereby affecting the diversity and stability of the gut microbiota. For example, AR may influence the immune-metabolic state of PCa patients, which in turn affects the composition of the microbiota and indirectly impacts the tumor’s immune microenvironment. Additionally, certain gut microbiota are closely related to the host’s hormone levels and can metabolize androgens, further regulating the metabolic environment of both the gut and the tumor [34]. Therefore, the relationship between gut microbiota and AR offers a new perspective for PCa research, especially in exploring the systemic effects of androgen signaling. Future investigations should focus on further elucidating the interaction between AR and gut microbiota, which will enhance our understanding of the pathological mechanisms of PCa and pave the way for developing new treatment strategies.

The influence of bacteria in the PCa microenvironment

Various bacteria, such as those from the gut microbiota, inhabit prostate tissue, leading scientists to investigate their role in PCa development [138, 139]. Bacteria’s presence in the PCa microenvironment can modulate host immune responses. Propionibacterium acnes and Escherichia coli are common in prostate tissue and closely linked to cancer development. Enrichment of Propionibacterium acnes in prostate tissue is linked to PCa development [140,141,142]. Moreover, Escherichia coli’s presence is closely tied to the PCa microenvironment. These bacteria can trigger host inflammatory responses, generating factors like interleukin-15 (IL-15). This process could inhibit PCa development by activating inflammatory pathways [142]. Additionally, inflammatory stress can induce tumor-associated epigenetic alterations, such as histone methylation, acetylation, and DNA methylation. The presence of bacteria in PCa could potentially influence the effectiveness of drug therapy. Several studies indicate that the presence of bacteria might disrupt drug distribution and metabolism within PCa tissue, thereby potentially impacting treatment efficacy [143,144,145]. This discovery is crucial for optimizing PCa treatment plans and gaining insights into personalized therapy [146]. Overall, bacteria in the PCa microenvironment could influence cancer development and treatment outcomes via multiple pathways. These findings are valuable not only for basic PCa research but also offer innovative directions for clinical medicine.

Impacts of viruses in the PCa microenvironment

Viruses in the PCa microenvironment have generated significant interest in the academic community [147]. Studies have demonstrated the presence of various viruses in this microenvironment [134, 148]. These viruses display intricate diversity within PCa tissues and affect the host organism at various biological levels. This virus can modulate PCa cell behavior through its impact on inflammation, angiogenesis, and apoptosis pathways [149]. In certain cases, within the PCa microenvironment, HPV infection has been detected, potentially influencing the proliferation and metastasis of PCa cells by modifying cell cycle regulation and manipulating host gene expression [150]. Specifically, the E6 and E7 proteins of HPV bind to p53 and retinoblastoma (Rb) proteins, respectively, disrupting their normal functions. This disruption subsequently affects cell cycle regulation and alters miRNA gene expression [134, 151]. HPV infection may offer a potential mechanism for PCa evasion by suppressing the host’s immune response, especially in recognizing and clearing tumor antigens [152]. Moreover, the presence of HHV is closely linked to PCa development and can affect tumor growth via immunosuppressive pathways. Complex interactions occur between these viruses and immune responses in the PCa microenvironment. HHV’s presence may reduce immune function, impairing the host’s response to cancer cells and promoting PCa development. The impact of viruses on PCa treatment is highly significant [135, 136]. In this context, thorough research into virus mechanisms in the PCa microenvironment is anticipated to yield fresh insights for future cancer research and provide more precise clinical management and treatment strategies.

Deciphering molecular pathways within the PCa microenvironment

Roles of signaling pathways

The intricate network of signaling pathways between PCa cells and their microenvironment profoundly influences the tumor’s behavior. Activation of the Wnt/β-catenin signaling pathway is closely linked to PCa cells proliferation, invasion, and metastasis [153,154,155]. Notably, this pathway’s activity is closely tied to inflammatory reactions in the PCa microenvironment, emphasizing its role in PCa development [156]. Precise regulation of the Wnt/β-catenin signaling pathway may present novel PCa treatment strategies. Conversely, regulation of the PI3K/Akt/mechanistic target of rapamycin kinase (mTOR) signaling pathway in the PCa microenvironment has garnered significant interest [157]. Overactivation of this pathway in PCa cells correlates with drug resistance and heightened inflammation in the microenvironment. Moreover, PI3K/Akt/mTOR pathway activation directly impacts PCa cells metabolic changes, creating a conducive environment for tumor cells survival [158]. A comprehensive study of this signaling pathway will elucidate cells survival mechanisms and treatment resistance in the PCa microenvironment. Research on PCa microenvironment signaling pathways not only reveals PCa initiation and progression mechanisms but also guides precision therapy development.

Wnt/β-catenin signaling pathway

The Wnt/β-catenin signaling pathway has multiple effects on the TME. Firstly, it significantly influences the number and function of cancer stem cells (CSCs) by regulating their self-renewal and differentiation processes [159]. Activation of this pathway is closely associated with maintaining and expanding PCa stem cells, impacting the tumor’s long-term survival and regeneration [160]. Secondly, activation of the Wnt/β-catenin signaling pathway also affects the polarity and migratory ability of PCa cells. By regulating cells adhesion and tight junctions, this signaling pathway influences PCa infiltration and metastasis [154, 161]. In terms of immune regulation, its activation not only inhibits anti-tumor immune responses but may also influence the quantity and activity of tumor-associated immune cells [162]. Abnormal activation of the Wnt/β-catenin signaling pathway may lead to PCa immune escape, reducing immune cells recognition and clearance of PCa cells, providing a potential mechanism for PCa immune escape [163]. Additionally, the Wnt/β-catenin signaling pathway contributes to the regulation of the ECM and angiogenesis in the PCa microenvironment [164]. Its activation may affect the synthesis and degradation of tumor-related stroma, altering the mechanical properties of the TME and cell-cell interactions. In terms of angiogenesis, activation of the Wnt/β-catenin signaling pathway is associated with factors related to PCa angiogenesis, potentially influencing the PCa’s blood supply by promoting the proliferation and migration of endothelial cells, affecting the PCa’s vascularization [165]. Moreover, researchers found through constructing datasets for primary PCa and benign prostate samples that the Wnt/β-catenin signaling pathway plays an important role, and the ubiquitin conjugating enzyme complex (UBE2C) may be a key node in this signaling pathway [166].

PI3K/Akt/mTOR signaling pathway

The PI3K/Akt/mTOR signaling pathway is a critical cellular signaling cascade that activates Akt protein phosphorylation via PI3K kinase activation, ultimately resulting in mTOR activation [167, 168]. This pathway directly regulates PCa cells metabolism, protein synthesis, and growth, exerting a direct impact on PCa development [169, 170]. Furthermore, the PI3K/Akt/mTOR signaling pathway is closely associated with inflammatory responses in the microenvironment of PCa. Aberrant activation of this pathway may lead to the release of inflammatory factors, inducing inflammatory responses in the TME, thereby affecting immune cells infiltration, activity, and significantly influencing PCa immune evasion [171]. Research has shown that the PI3K/Akt signaling pathway can promote the release of exosomes and upregulation of PD-L1 expression in PCa cells, which greatly alters the immunosuppressive state and affects macrophage polarization [172]. Additionally, the PI3K/Akt/mTOR signaling pathway directly participates in regulating angiogenesis by modulating VEGF and other molecule expressions, thus affecting PCa vasculature and impacting PCa growth and metastasis [83, 173]. The PI3K/Akt/mTOR signaling pathway also contributes significantly to the development of treatment resistance in PCa. Research indicates that as PCa progresses to a resistant stage, it may shift from relying on AR signaling to dependence on the PI3K/Akt/mTOR pathway. Hyperactivation and dysregulation of this pathway are linked to increased tumor aggressiveness [174, 175]. Thus, targeting the PI3K/Akt/mTOR pathway may offer a promising therapeutic strategy to overcome resistance in PCa. Therefore, attaining a thorough comprehension of the PI3K/Akt/mTOR signaling pathway’s involvement in the microenvironment of PCa is anticipated to unveil novel insights and therapeutic targets for PCa management.

Cytokines and regulatory factors

The TNF family members play diverse roles in immune response regulation and cells survival [176]. Studies show tumor necrosis factor-α (TNF-α) is overexpressed in PCa, closely linked to tumor development and microenvironmental inflammation [177, 178]. Further investigation is needed to determine the specific roles of tumor necrosis factor-β (TNF-β) and factor associated suicide ligand (FasL) in the PCa microenvironment. Precise cytokine regulation can significantly impact tumor growth, infiltration, and metastasis. Transcription factor regulation is crucial for tumor development in the PCa microenvironment. NF-κB is a critical transcription factor within the regulatory network of PCa [179, 180]. Thus, studying NF-κB’s role and regulatory network in the PCa microenvironment is crucial for understanding tumor pathogenesis and identifying new therapeutic targets.

TNF family

The TNF family’s extensive involvement in the PCa microenvironment has significant implications for tumor development and treatment response [181]. This is a family of multiple cytokines that play important roles in various biological processes such as cell proliferation, differentiation and apoptosis, immune regulation, inflammatory response, angiogenesis, and invasion. Certain TNF family members can directly regulate PCa cells proliferation by activating some specific signaling pathways. TNF-α is an important member of the TNF superfamily, with a wide range of biological functions and important roles in PCa [182]. TNF-α can induces proliferation and invasion of PC3 cells through activation of PI3K/Akt and NF-κB signaling pathways [183]. Moreover, some TNF family members regulate cells apoptosis, affecting the survival status of PCa cells [184]. Fas/APO-1/CD95 is a member of the TNF receptor superfamily, which can induce tumor cell apoptosis. In the PCa microenvironment, the Fas signaling pathway has complex interactions with other important tumor signaling pathways, which further regulates the survival, proliferation, and apoptosis of PCa cells [185]. Research has shown that Fas induces apoptosis in DU145 cells by activating JNK and MAPK, which in turn affect the downstream BCL2 family [186]. These findings highlight the critical role of the TNF family as key regulators of cells fate in the PCa microenvironment. Recent studies have shown that some TNF family members play crucial regulatory roles in immune cells infiltration and activation [187, 188]. These members such as TNF-α and TNF-related apoptosis-inducing ligand (TRAIL) may influence PCa’s immune evasion by modulating the activity of immune cells like T cells and NK cells [189, 190]. Additionally, the TNF family regulates the expression of immune checkpoint molecules, affecting PCa’s sensitivity to immunotherapy [191]. TNF family members also play a significant role in angiogenesis, PCa invasion, and metastasis in the PCa microenvironment. In the TME of PCa, TRAIL plays an important role, not only affecting immunity, but also regulating invasion and metastasis [192, 193]. Recent research shows that certain TNF family members directly affect PCa’s blood supply and growth by regulating vascular endothelial cells function and angiogenesis-related signaling pathways [194, 195]. These insights deepen our understanding of the TNF family’s role in PCa’s microenvironment.

Transcription factors

Transcription factors in the PCa microenvironment profoundly regulate PCa development through multiple mechanisms [196, 197]. NF-κB participates in regulating inflammation and modulates key processes such as cells survival, proliferation, and invasion in PCa occurrence and progression [198, 199]. Activity regulation of NF-κB may be closely associated with inflammation status, cell-cell interactions, and immune escape in the PCa microenvironment. In terms of inflammatory status in the PCa microenvironment, NF-κB can regulate the expression of various pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β [200, 201]. The excessive activation of NF-κB can maintain and exacerbate the inflammatory state, and inhibiting the activity of NF-κB may be a potential therapeutic strategy that can slow down or prevent the progression of PCa. In addition, NF-κB can affect the escape of tumor cells from the immune system. It can promote the expression of PD-L1, and the binding of PD-L1 to programmed cell death protein-1 (PD-1) receptor can inhibit the activity of T cells, thereby helping tumor cells escape immune surveillance [32, 202]. Furthermore, transcription factors regulate PCa cells surface antigen expression, affecting PCa cells recognition efficiency by the immune system and modulating PCa immune escape [203]. Certain transcription factors can interact with proteins related to cells cycle regulation, such as cyclin-dependent kinases (CDKs) and cyclin-dependent kinase inhibitors (CKIs), to regulate PCa cells cycle progression [204, 205]. Transcription factors also directly regulate DNA replication and chromosome segregation, thus directly influencing cells proliferation [206]. Specific transcription factors can directly intervene in immune cells function in the PCa immune microenvironment [207,208,209]. This involves transcription factors regulating immune cells surface receptors, co-stimulatory molecules, and cytokines, impacting immune cells activation, proliferation, and effector functions. Specific transcription factors directly regulate genes related to angiogenesis, including VEGF and basic fibroblast growth factor (bFGF). This regulation influences oxygen and nutrient supply to PCa cells, affecting PCa growth and invasion [65, 210, 211]. In summary, transcription factors profoundly impact PCa microenvironment regulation through direct involvement in cells cycle regulation, immune modulation, angiogenesis, and nutrient supply, among other mechanisms.

Microbiota-driven molecular pathways

Bacterial mediated

Recent research indicates that the composition of gut bacteria can impact the efficacy of tumor immunotherapy [212]. In germ-free or symbiotic eradication conditions of the gut microbiota, the efficacy of immunotherapy drugs like PD-L1 inhibitors are significantly reduced in various animal tumor models. Human study has also shown that abnormal gut microbiota composition can lead to immune checkpoint inhibitor (ICI) resistance in epithelial cells [213]. In their study involving 23 patients with metastatic castration-resistant prostate cancer (mCRPC) receiving enzalutamide treatment, Peiffer et al. used fecal sequencing of gut microbiota. They found that although there was no significant difference in gut microbiota diversity between responders and non-responders, responders had higher levels of Streptococcus salivarius in their feces compared to non-responders. Oral Streptococcus salivarius levels were not linked to response status [214]. Furthermore, muciniphila has shown a correlation with immune therapy response in PCa, exhibiting increased abundance in the fecal samples of PD-1 inhibitor responders. Conversely, its levels are reduced in mCRPC. This suggests a novel approach to boosting immune therapy response through fecal transplant from responders, although more research is needed to confirm its clinical viability [215]. Recent studies have shown that the bacterium Ruminococcus is significantly enriched in patients with CRPC and is associated with phospholipid metabolism [216, 217]. These findings are crucial for optimizing PCa treatment plans and understanding the potential for personalized therapy. Furthermore, current research on immune therapy in PCa and the microbiota predominantly centers on gut microbiota, with few studies investigating microbiota in the prostate or urogenital tract, which could also influence immune therapy effectiveness in PCa. Thus, future research should investigate the connection between these microbiota and immune therapy.

Recent studies have indicated that gut commensal bacteria may play a significant role in the production and metabolism of androgens within the body, even in patients undergoing ADT. For example, some research has found that certain gut microbiota can produce androgens by metabolizing precursor substances, thereby affecting the host’s hormone levels and the immune microenvironment of tumors [218, 219]. The androgen-producing capabilities of these bacteria may significantly impact the effectiveness of ADT and the progression of the disease. Therefore, exploring the mechanisms of androgen production and secretion by gut microbiota in the context of ADT is crucial for a comprehensive understanding of their influence on PCa. Future research should focus on elucidating the role of gut commensal bacteria in androgen metabolism in ADT patients to reveal their potential impact on PCa development. This will not only enhance our understanding of the pathological mechanisms of PCa but may also provide important insights for the development of new therapeutic strategies.

Virus mediated

HPV

HPV is a group of viruses with pleomorphic characteristics that can infect various parts of the human body, such as the skin, reproductive tract, urethra, and respiratory tract, leading to tissue proliferation, verrucous epithelial lesions, malignant tumors, etc [220]. According to statistics from Russo et al. in 2018, there were 98 studies from 1990 to 2015 on the association between HPV infection and the incidence of PCa, involving over 9,000 participants, with 3,939 individuals in the experimental group and 5,683 in the control group [221]. This study primarily focused on the association between HPV-16 and HPV-18 infections and PCa. Meta-analysis results showed that HPV-16 infection increased the average odds ratio (OR = 1.61) of PCa risk, while HPV-18 infection had a relatively lower average odds ratio (OR = 1.20) of PCa risk. From the data results, the association between HPV-16 and PCa appears more pronounced, while the relationship between HPV-18 and PCa may require more experimental data for verification.

EBV

EBV was initially discovered and isolated in biopsy samples from Burkitt’s Lymphoma patients in 1964. Studies have demonstrated notable differences in anti-apoptotic factors (e.g., survivin and Bcl-2) and tumor suppressor factors (e.g., p53 and Rb) among EBV-infected PCa samples, highlighting the potential influence of EBV on PCa development [222, 223].

HHV

The HHV comprises herpes simplex virus type-1 (HSV-1) and herpes simplex virus type-2 (HSV-2), representing typical herpesvirus types [224]. Acute herpes simplex virus (HSV) infection causes vesicular dermatitis and can lead to inflammations and reproductive system infections. Studies show that the oncolytic HSV, combined with the PI3K inhibitor BKM120, synergistically enhances the eradication of PCa stem-like cells [225]. Major discoveries about HSV and new PCa therapies present novel opportunities for PCa diagnosis and treatment, rendering related research a recent hot topic [226].

The role of bacteria and viruses in the PCa microenvironment entails intricate molecular mechanisms significantly influencing tumor progression and treatment response. Uncovering these mechanisms mediated by bacteria and viruses aids in enhancing the comprehension of PCa pathophysiological processes, offering fresh perspectives for designing future targeted therapy and immunotherapies (Table 1).

Table 1 Extrinsic factors in TME cells and internal factors in tumor cells and their effects

Significance of PCa microenvironment in clinical practice

PCa diagnosis

In the field of PCa diagnosis, research on microenvironmental markers primarily involves multiple aspects, including genetic variations, ECM, extracellular vesicles, and metabolites [227]. Identifying specific TME biomarkers is crucial in diagnosing PCa. These biomarkers encompass various types, including ECM proteins (e.g., Collagen type VII, MMP-1, MMP-2, MMP-7, MMP-9, MT1-MMP, CUL4B) indicating tumor invasiveness [228,229,230]; CSCs markers (e.g., BTF3, Skp2, BMI1, Sox2, ALDH1A1, ALDH1A3, ACK1, TNK2) influencing PCa recurrence and treatment resistance [231,232,233,234]; transcription factors (e.g., NF-κB) associated with PCa initiation and progression [235]; IL (e.g., IL-6, IL-8) pivotal in PCa inflammation and progression [236, 237]; growth factors (e.g., VEGF, FGF1, FGF2, FGF6, FGF8, TGF-B, PDGF) critical in tumor immune evasion and microenvironment regulation [238,239,240]; immunomodulators (e.g., PTX3, COX-2) aiding in assessing tumor immune escape and potential targets for ICI therapy [241, 242]; angiogenic stimulators (e.g., Ang-1, EGF, TGF-α) significant in modulating the TME and promoting growth [243, 244]; chemokines (e.g., CXCL12) pivotal in tumor cell migration and metastasis [245]; and glycoproteins (e.g., THBS4) promoting tumor progression via the PI3K/Akt pathway [246]. Detecting these biomarkers aids in early PCa diagnosis and predicts disease progression and patient prognosis, offering crucial insights for personalized treatment strategies. In-depth study of genetic variations, precise analysis of specific genes in the PCa microenvironment is achieved. These studies have found that mutations or copy number variations in specific genes may exhibit significant specificity, becoming one of the indicators of early carcinogenesis. The application of high-throughput sequencing technology enables us to comprehensively analyze the genetic heterogeneity of cells populations in the microenvironment, providing a profound genetic basis for the selection of potential biomarkers [247]. Secondly, the protein components in the ECM are also a key aspect of microenvironmental marker research [7]. The interaction between PCa cells and the surrounding matrix triggers changes in matrix components, including protein expression and modification. Through an in-depth study of these matrix proteins’ changes, specific markers related to the PCa microenvironment can be discovered [248]. These proteins may involve key biological processes such as cells adhesion, signal transduction, and matrix remodeling, providing detailed information on the carcinogenesis process. Extracellular vesicles, as a medium for extracellular communication, also serve as a potential source of microenvironmental markers [249]. The nucleic acids, proteins, and other molecules carried by these vesicles may change the PCa microenvironment. By analyzing the composition of extracellular vesicles, specific markers related to cancer can be identified, providing new biological markers for diagnosis. Changes in metabolites are also an important direction in microenvironmental marker research [227, 250]. Metabolomic methods can reveal abnormalities in specific metabolic pathways in the PCa microenvironment, thus identifying metabolites associated with cancer. These metabolites may involve key biological processes such as lipid metabolism, sugar metabolism, and amino acid metabolism, providing more precise biological markers for early diagnosis [251, 252]. Therefore, through in-depth study of these microenvironmental markers, more accurate and detailed molecular-level information can be provided for the early diagnosis of PCa, providing strong support for personalized treatment and clinical decision-making (Fig. 6).

Fig. 6
figure 6

Exploring the PCa microenvironment: new advances in diagnosis. The diagnosis of the PCa microenvironment involves various biomarkers, including growth factors, pro-angiogenic factors, immune mediators, IL, chemokines, ECM components, CSCs, glycoproteins, and transcription factors

PCa therapeutic strategies

In the section on therapeutic strategies for PCa, we highlight the importance of immunotherapy and stress the necessity of developing comprehensive treatment plans that consider the characteristics of the TME. However, the limited or absent responses seen in current PCa treatment strategies are mainly due to several factors. Firstly, the complexity and heterogeneity of the TME have a significant impact on treatment outcomes. Particularly, diverse populations of immunosuppressive cells like Treg cells and MDSCs in the PCa microenvironment hinder effective anti-tumor immune responses through the secretion of suppressive cytokines such as TGF-β and IL-10 [253, 254]. Secondly, PCa cells often exhibit high levels of the immune checkpoint molecule PD-L1, which binds to the PD-1 receptor, suppressing T cell function and enabling evasion from immune surveillance [255]. Specifically, the PD-L1/PD-1 pathway downregulates T cell activation and proliferation, reducing their cytotoxic activity and thereby diminishing the immune system’s ability to target PCa cells. This mechanism allows PCa cells to escape immune surveillance and continue to grow and metastasize within the TME. Studies have shown that PCa cells upregulate PD-L1 expression, significantly enhancing their immune evasion capabilities. This phenomenon is particularly evident in PCa patient tumor tissues, where high PD-L1 expression correlates closely with tumor progression and immune evasion [32, 256, 257]. Additionally, PCa cells can secrete various immune-suppressive factors, such as IL-10 and TGF-β, which further inhibit T cell function and promote the expansion of immune-suppressive cell populations, such as regulatory T cells. Overall, PCa cells alter immune surveillance through upregulation of PD-L1 and other immune-suppressive mechanisms, thereby facilitating persistent tumor growth and metastasis. Additionally, the presence of CSCs and the expression of multidrug resistance genes like ATP binding cassette subfamily G member 2 (ABCG2) and ATP binding cassette subfamily B member 1 (ABCB1) substantially reduce the effectiveness of current treatment strategies, as these CSCs have increased tolerance to traditional therapies and significant potential for recurrence [258]. Developing more effective combination therapy strategies presents several challenges. Firstly, optimizing combinations of various treatment modalities like immunotherapy, targeted therapy, and radiation therapy is crucial to maximize therapeutic effectiveness. For example, combining immune checkpoint inhibitors (ICIs) with anti-angiogenic therapy can overcome the limitations of single-agent therapy [259] (Fig. 7a). Secondly, overcoming immunosuppressive factors in the TME by targeting the TGF-β signaling pathway or using Treg cells inhibitors is vital to enhance anti-tumor immune responses [260, 261]. Additionally, identifying and targeting key signaling pathways such as PI3K/Akt and MAPK can enhance immune cell function while inhibiting tumor cell growth and survival [262]. Finally, developing drug delivery systems that can penetrate the barriers of the TME, such as nanoparticles and oncolytic viruses, is crucial for enhancing therapeutic effectiveness [263, 264] (Fig. 7b).

Fig. 7
figure 7

Exploring the PCa microenvironment: new advances in treatment. A Immunotherapy strategies currently emphasize the use of PD-L1/PD-1 inhibitors in conjunction with anti-VEGF and anti-PD-L1 therapies, highlighting their increasing significance. B The development of drug delivery systems like nanoparticles and oncolytic viruses, capable of breaching the TME barrier, may enhance therapy effectiveness. c. Ongoing exploration centers on microbial therapy strategies targeting viruses and bacteria

Immunotherapy has become widely recognized in recent years for its distinct mechanisms of action. However, its applicability and efficacy within the TME remain a complex and worthy topic for in-depth research [265]. To precisely tailor immunotherapy regimens, there is an urgent need to gain a deeper understanding of the PCa microenvironmental characteristics [266]. Furthermore, research should focus on the combination of immunotherapy with other treatment modalities to enhance therapeutic efficacy and reduce adverse reactions in patients. Targeting drugs against the PCa microenvironment has become one of the focal points in current cancer treatment research. The complexity of the PCa microenvironment underscores the crucial importance of designing targeted drugs. By delving into the interactions between PCa cells and their surrounding environment, it is hoped that new therapeutic targets will be discovered, leading to the development of more targeted and efficient drugs [267,268,269,270]. For example, drugs inhibiting PCa angiogenesis or targeting PCa-associated immune checkpoint molecules could potentially become integral components of future treatment strategies. The consideration of microbiota in treatment strategies is also at the forefront of current research. The interaction between gut microbiota and the immune system has profound implications on PCa development and treatment response [138]. Thus, modulating gut microbiota composition could impact patients’ response to immunotherapy (Fig. 7c). For this purpose, a thorough comprehension of the roles and interconnections among various microbiota is essential for achieving targeted modulation of microbiota composition in therapeutic approaches [17]. In conclusion, insights into therapeutic strategies from the microenvironment need to comprehensively cover the potential applications of immunotherapy, drug research targeting the PCa microenvironment, and considerations of microbiota in treatment strategies, these three key areas. Through in-depth research in these aspects, it is expected that more personalized and efficient treatment methods will be discovered, leading to better clinical outcomes for PCa patients. This series of studies will provide new directions and concepts for the future development of the field of cancer treatment.

PCa clinical implications and translational potential

This review summarizes various components of the PCa microenvironment, such as immune cells, the vascular system, stromal cells, and microbiota. However, translating these findings into clinical practice requires further attention and in-depth research. Current studies mainly focus on the basic biological properties and functions of the components within the PCa microenvironment. Nonetheless, many challenges remain in effectively regulating these microenvironments in clinical settings to improve therapeutic outcomes.

Tumor cells often recruit immune cells to the tumor site to combat foreign pathogens and infections [271]. In the PCa microenvironment, however, the functions of immune cells like T cells and NK cells are often suppressed, impairing the anti-tumor immune response. Current research focuses on restoring the activity of these immune cells, with the most promising strategies being inhibitors targeting the PD-1/PD-L1 immune checkpoint. Pembrolizumab, as demonstrated in the KEYNOTE-199 trial, has shown efficacy in patients with advanced PCa. Pembrolizumab, combined with other treatments, is currently in phase III clinical trials to further improve response rates and patient survival [272,273,274]. Additionally, the cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) inhibitor ipilimumab and the poly ADP-ribose polymerase (PARP) inhibitor olaparib are undergoing phase III clinical trials for treating mCRPC and docetaxel-resistant diseases [275,276,277].

The vascular system is crucial in the PCa microenvironment. VEGF promotes tumor growth and nourishment. Anti-VEGF drugs, including bevacizumab, apatinib, pazopanib, AZD2171, and sunitinib, have demonstrated efficacy in treating PCa. Ongoing phase II clinical trials are evaluating bevacizumab for CRPC, with preliminary results showing significant potential in delaying disease progression [278]. Apatinib and pazopanib have shown promising results in phase I/II clinical trials for mCRPC [279, 280]. Vascular endothelial growth factor receptor-1 (VEGFR-1) and vascular endothelial growth factor receptor-2 (VEGFR-2) inhibitors, such as AZD2171, have shown good therapeutic effects in phase I trials for CRPC [281]. The VEGFR-2 inhibitor sunitinib is in phase II clinical trials for mCRPC [282]. Additionally, nintedanib, a drug that inhibits FGF signaling pathways, has shown potential in reducing tumor growth and metastasis in phase II clinical trials. AT-101, a lactate dehydrogenase (LDH) inhibitor, binds to and inhibits the anti-apoptotic function of Bcl-2, effectively stimulating pro-apoptotic proteins. It has shown efficacy in PCa treatment and is currently in phase II trials for metastatic hormone-sensitive prostate cancer (mHSPC) [283,284,285].

Stromal cells provide critical support within the PCa microenvironment by secreting various cytokines and growth factors, promoting tumor growth and metastasis. New therapeutic strategies targeting stromal cells are being developed [286]. For example, tranilast, which inhibits the production of transforming growth factor-β1 (TGF-β1) by bone stromal cells, has shown effectiveness against metastatic CRPC. It is currently in phase II clinical trials to assess its safety and efficacy in PCa treatment [287]. Additionally, AT-101, which enhances the immune surveillance and clearance abilities of senescent fibroblasts, is in phase II clinical trials for CRPC treatment [288, 289].

The role of the microbiome in PCa progression is gaining attention. Research suggests that gut microbiota dysbiosis may influence PCa occurrence and development through immunomodulatory mechanisms. New microbiome modulation approaches to intervene in PCa are being explored. Preliminary data indicate that regulating the gut microbiota can enhance the immune response and improve treatment outcomes [290]. For example, the antibiotic cocktail therapy ABX, which reduces short-chain fatty acid (SCFA) production by gut microbiota and lowers insulin-like growth factor-1 (IGF-1) levels, is in phase III clinical trials for CRPC and shows improved treatment outcomes [291, 292]. Additionally, vaccination plays a significant role in PCa clinical treatment. The peptide viral vaccine PROSTVAC-V/F, which inhibits PD-1, has shown efficacy in inhibiting the progression of metastatic CRPC and is now in phase III clinical trials [293, 294]. The prostate acid phosphatase DNA vaccine (pTVG-HP) has shown efficacy in phase II trials for hormone-sensitive prostate cancer (HSPC) [295]. Sipuleucel-T, a monocyte-based cancer vaccine, is the only FDA-approved therapeutic vaccine for PCa and is a treatment option for asymptomatic and minimally symptomatic patients in North America. It is currently in phase II trials for CRPC [296, 297] (Table 2).

Table 2 PCa clinical implications and translational potential

Moreover, identifying predictive biomarkers for immunotherapy response is critical for improving the diagnosis and treatment of PCa. Current biomarkers that may influence the efficacy of immunotherapy in PCa include genetic-based markers such as tumor mutational burden (TMB), microsatellite instability/mismatch repair (MSI/MMR), and cyclin-dependent kinase 12 (CDK12) mutations, along with cell type proportion markers like the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) [298,299,300,301].

In recent years, TMB has gained significant attention as a biomarker in ICI research. Research indicates that PCa patients with high TMB are more likely to benefit from ICI therapy. Tumors with high TMB typically express numerous abnormal proteins, which are displayed on the surface of tumor cells and recognized by immune cells, thereby triggering an immune response [302]. A large retrospective study in advanced PCa showed that patients with TMB ≥ 10 mutations per megabase (mt/Mb) experienced prolonged time to next treatment and significantly improved overall survival following ICI therapy, compared to those receiving taxane chemotherapy [303].

Mismatch repair deficiency (dMMR) results from the loss of any mismatch repair protein, leading to the accumulation of errors during DNA replication and ultimately causing MSI. Tumors with high microsatellite instability (MSI-H)/dMMR often show increased T-cell infiltration, making them more responsive to ICIs [304, 305]. In a study by Abida et al., 6 out of 11 mCRPC patients with MSI-H/dMMR exhibited a significant decline in prostate-specific antigen (PSA) levels following ICI treatment [306].

Loss of CDK12 disrupts DNA damage repair, leading to increased gene fusions, a higher neoantigen load, and enhanced T-cell infiltration. Several studies have reported that PCa patients with CDK12 mutations are more likely to respond to ICIs. Wu et al. observed that 2 out of 4 mCRPC patients with biallelic CDK12 inactivation responded positively to PD-1 inhibitor monotherapy [307]. Similarly, Antonarakis et al. found that 3 out of 9 mCRPC patients with CDK12 mutations achieved partial responses after treatment with nivolumab [308].

A meta-analysis of 7,228 PCa patients found that elevated NLR and PLR were associated with poor prognosis, suggesting their potential as prognostic biomarkers. However, the mechanisms underlying the relationship between elevated NLR/PLR and adverse outcomes in PCa remain unclear and require further investigation [309].

Conclusion and perspective

PCa stands out as one of the prevailing male malignancies, commanding widespread attention due to its profound impact on public health. As medical technology advances at a rapid pace, the intricate landscape of the PCa microenvironment has emerged as a focal point for comprehensive investigation. Delving into the microenvironment of PCa unveils a labyrinthine network system characterized by its complexity and multi-level interactions. Comprising immune cells, angiogenic factors, stromal cells, and an array of molecular signaling pathways, among other constituents, this milieu offers a rich tapestry of avenues for unraveling the mechanisms underlying PCa pathogenesis and progression. The microenvironment of PCa serves as a dynamic arena wherein myriad cellular and molecular players orchestrate a delicate balance between tumor suppression and promotion. Understanding the interplay between these elements holds promise for the development of novel therapeutic modalities and preventive strategies aimed at mitigating the burden of PCa. Notably, insights gleaned from dissecting the PCa microenvironment have the potential to revolutionize existing paradigms in cancer treatment, fostering tailored approaches that capitalize on the unique vulnerabilities of the tumor milieu. While the precise determinants driving the high incidence of PCa remain incompletely elucidated, a constellation of factors, including environmental exposures, genetic predispositions, and lifestyle variables, have been implicated in shaping the risk landscape of this disease. Unraveling the intricate interplay between these factors and their influence on the PCa microenvironment represents a critical frontier in the quest to curtail the burgeoning burden of this disease. In essence, the exploration of the PCa microenvironment transcends the confines of traditional oncological research paradigms, heralding a new era characterized by precision medicine and personalized interventions tailored to the unique molecular landscape of individual tumors. By deciphering the intricacies of the PCa microenvironment, researchers stand poised to forge innovative therapeutic strategies that hold the promise of transforming the management of this formidable disease entity.

This review comprehensively summarized the role of the microenvironment in PCa, focusing on the specific background of PCa, and providing in-depth analysis of immune cells, angiogenesis, and molecular mechanisms. This not only helps to comprehensively understand the pathogenesis of PCa but also provides rich ideas for future treatments. Based on this, further exploration of the unique characteristics of the PCa microenvironment is warranted. The role of immune cells in the PCa microenvironment, including different subsets of T lymphocytes and their functions, as well as the crucial role of APCs, is carefully analyzed. The relationship between angiogenesis and PCa, including the regulation of the VEGF family and the complex interaction between endothelial cells and cancer cells, reveals the delicate balance between PCa growth and blood supply. The functions of stromal cells, especially the synergistic effects of CAFs and stromal cells, and the impact of ECM changes on PCa development, are elaborated in this review, focusing on molecular mechanisms research. The role of signaling pathways in the PCa microenvironment, with particular emphasis on the regulatory mechanisms of the Wnt/β-catenin and PI3K/Akt signaling pathways, is explored. The importance of cytokines and regulatory factors, especially the role of the TNF family and transcription factors in the regulation of the PCa microenvironment, uncovers mysteries at the molecular level. An in-depth exploration of the potential clinical applications of the PCa microenvironment reveals the impact of the microenvironment on PCa diagnosis, including the potential application of microenvironmental biomarkers, providing clinicians with more precise diagnostic tools. Insights into treatment strategies from the microenvironment, including the potential application of immunotherapy and targeted drug research on the PCa microenvironment, provide new directions for future treatments. Reviewing the challenges in the current research field of PCa microenvironment and the development of new technological methods, it becomes evident that addressing technical limitations and theoretical challenges necessitates collaborative efforts to achieve breakthroughs.

We have noticed that there are still some challenges in the clinical translation and application of PCa microenvironment research. Although we have mentioned several microenvironment biomarkers, their accuracy, specificity, and clinical predictive value still need further validation in clinical practice. Additionally, a deeper understanding of the interactions among various biomarkers in the microenvironment needs to be strengthened, which will help reveal more details about the complexity of the TME. Firstly, in current PCa microenvironment research, we must recognize that there are some challenges. For example, in terms of clinical translation, although we have identified multiple microenvironment biomarkers, their accuracy and specificity have not been fully validated in clinical practice. Furthermore, the existing research lacks a thorough understanding of the interactions among various biomarkers in the microenvironment, leading to a need for a more comprehensive understanding of the complexity of the TME. In response to these challenges, we propose several future research directions: Establishment of biomarker combination models: develop combination models for different PCa subtypes and clinical stages, combining multiple microenvironment biomarkers to improve diagnostic and predictive accuracy. Application of bioinformatics and artificial intelligence in microenvironment analysis: utilize bioinformatics and artificial intelligence technologies to delve into microenvironment data, discover new biomarkers, signaling pathways, and their clinical significance. Research on microenvironment-regulated treatment strategies: explore treatment strategies targeting the microenvironment, such as targeted therapy for specific biomarkers, immunomodulatory therapy, etc., to achieve more precise personalized treatment. Exploration of precise treatment strategies for microenvironment regulation: explore precise treatment strategies targeting the microenvironment, such as targeted therapy for specific biomarkers, immunomodulatory therapy, etc., to achieve personalized treatment and improve treatment effectiveness. Multi-center large-sample clinical validation studies: conduct multi-center, large-sample clinical validation studies to verify the feasibility and effectiveness of new biomarkers or models in clinical practice, thereby accelerating their clinical application. Precision medicine perspective: advocate starting from the perspective of precision medicine, combining personalized treatment strategies and microenvironment characteristics, develop biomarker combination models suitable for different patients, and apply them in clinical practice. Prospective clinical research: we advocate for prospective, multi-center clinical research to validate the feasibility and effectiveness of new biomarker combination models in different clinical scenarios, providing more reliable evidence for personalized treatment. Exploring these future research directions will provide a more comprehensive and in-depth understanding of PCa microenvironment research, promote the clinical application of microenvironment biomarkers, and lay a more solid scientific foundation for personalized treatment and precision medicine in PCa.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

AAC:

Amino acid chain

ABCB1:

ATP binding cassette subfamily B member 1

ABCG2:

ATP binding cassette subfamily G member 2

ACK1:

Activated CDC42 kinase-1

ADT:

Androgen deprivation therapy

Akt:

Protein kinase B

ALDH1A1:

Aldehyde dehydrogenase 1A1

ALDH1A3:

Aldehyde dehydrogenase 1A3

Ang-1:

Angiopoietin-1

APCs:

Antigen-presenting cells

AR:

Androgen receptors

ATG5:

Autophagy-related gene 5

bFGF:

Basic fibroblast growth factor

BMI1:

B lymphoma Mo-MLV insertion region 1

BTF3:

Basal transcription factor 3

CAFs:

Cancer-associated fibroblasts

CD28:

Cluster of differentiation 28

CD31:

Cluster of differentiation 31

CD40:

Cluster of differentiation 40

CD40L:

Cluster of differentiation 40 ligand

CD80:

Cluster of differentiation 80

CD86:

Cluster of differentiation 86

CDKs:

Cyclin-dependent kinases

CDK12:

Cyclin-dependent kinase 12

CKIs:

Cyclin-dependent kinase inhibitors

COX-2:

Cyclooxygenase-2

CRPC:

Castration-resistant prostate cancer

CSCs:

Cancer stem cells

CTL:

Cytotoxic CD8+ T lymphocyte

CTLA-4:

Cytotoxic T lymphocyte-associated antigen-4

CUL4B:

Cullin 4B

CXCL-12:

C-X-C motif chemokine ligand-12

DCs:

Dendritic cells

dMMR:

Mismatch repair deficiency

EBV:

Epstein-barr virus

ECM:

Extracellular matrix

EGF:

Epidermal growth factor

EMT:

Epithelial-mesenchymal transition

Fas:

Factor associated suicide

FasL:

Factor associated suicide ligand

FGF:

Fibroblast growth factor

FGF1:

Fibroblast growth factor 1

FGF2:

Fibroblast growth factor 2

FGF6:

Fibroblast growth factor 6

FGF8:

Fibroblast growth factor 8

FGFR:

Fibroblast growth factor receptor

GPCRs:

G protein-coupled receptors

HHV:

Human herpes virus

HPV:

Human papilloma virus

HSV:

Herpes simplex virus

HSV-1:

Herpes simplex virus type-1

HSV-2:

Herpes simplex virus type-2

ICI:

Immune checkpoint inhibitor

ICIs:

Immune checkpoint inhibitors

IFN-γ:

Interferon-γ

IGF-1:

Insulin-like growth factor-1

IL:

Interleukin

IL-2:

Interleukin-2

IL-6:

Interleukin-6

IL-7:

Interleukin-7

IL-8:

Interleukin-8

IL-10:

Interleukin-10

IL-15:

Interleukin-15

IL-23:

Interleukin-23

IL-30:

Interleukin-30

LDH:

Lactate dehydrogenase

M2-MФ:

M2-macrophages

MAPK:

Mitogen-activated protein kinase

mCRPC:

Metastatic castration-resistant prostate cancer

mDCs:

Myeloid dendritic cells

MDSCs:

Myeloid-derived suppressor cells

MHC-I:

Major histocompatibility complex-I

MHC-II:

Major histocompatibility complex-II

MMP-14:

Matrix metalloproteinase-14

MMP-2:

Matrix metalloproteinase-2

MMP-3:

Matrix metalloproteinase-3

MMP-7:

Matrix metalloproteinase-7

MMP-9:

Matrix metalloproteinase-9

MMPs:

Matrix metalloproteinases

MMR:

Mismatch repair

MSI:

Microsatellite instability

MSI-H:

High microsatellite instability

MT1-MMP:

Membrane type 1-matrix metalloproteinase

mTOR:

Mechanistic target of rapamycin kinase

MUC1-C:

Mucin 1-C

mt/Mb:

Mutations per megabase

NFs:

Normal fibroblasts

NF-κB:

Nuclear factor-kappa B

NLR:

Neutrophil-to-lymphocyte ratio

NK:

Natural killer

PARP:

Poly ADP-ribose polymerase

PCa:

Prostate cancer

PD-1:

Programmed cell death protein-1

pDCs:

Plasmacytoid dendritic cells

PDGF:

Platelet-derived growth factor

PD-L1:

Programmed cell death-ligand 1

PECAM-1:

Platelet endothelial cell adhesion molecule-1

PGE2 :

Prostaglandin E2

PI3K:

Phosphoinositide 3-kinase

PLGF:

Placental growth factor

PLR:

Platelet-to-lymphocyte ratio

PSA:

Prostate-specific antigen

pTVG-HP :

Prostate acid phosphatase DNA vaccine

PTX3:

Pentraxin 3

Rb:

Retinoblastoma

RBCs:

Red blood cells

SCFA:

Short-chain fatty acid

Skp2:

S-phase kinase associated protein 2

SNs:

Sentinel lymph nodes

Sox2:

Sex determining region Y-box 2

TAMs:

Tumor-associated macrophages

TCMs:

Traditional Chinese medicines

TCR:

T cell receptor

TGF-α:

Transforming growth factor-α

TGF-β:

Transforming growth factor-β

TGF-β1:

Transforming growth factor-β1

Th:

Helper CD4+ T

THBS4:

Thrombospondin 4

TIMo:

Tumor-inflammatory monocytes

TLR:

Toll like receptor

TME:

Tumor microenvironment

TMB:

Tumor mutational burden

TNF:

Tumor necrosis factor

TNF-α:

Tumor necrosis factor-α

TNF-β:

Tumor necrosis factor-β

TNK2:

Tyrosine kinase non receptor 2

TRAIL:

TNF-related apoptosis-inducing ligand

Treg:

Regulatory CD4+ T

TYR:

Thyrosinase

UBE2C:

Ubiquitin conjugating enzyme complex

VCAM:

Vascular cell adhesion molecule

VEGF:

Vascular endothelial growth factor

VEGF-A:

Vascular endothelial growth factor-A

VEGF-B:

Vascular endothelial growth factor-B

VEGF-C:

Vascular endothelial growth factor-C

VEGF-D:

Vascular endothelial growth factor-D

VEGFR:

Vascular endothelial growth factor receptor

VEGFR-1:

Vascular endothelial growth factor receptor-1

VEGFR-2:

Vascular endothelial growth factor receptor-2

References

  1. Eeles R, Goh C, Castro E, Bancroft E, Guy M, Al Olama AA, Easton D, Kote-Jarai Z. The genetic epidemiology of prostate cancer and its clinical implications. Nat Rev Urol. 2014;11:18–31.

    Article  PubMed  CAS  Google Scholar 

  2. Qiu H, Cao S, Xu R. Cancer incidence, mortality, and burden in China: a time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in 2020. Cancer Commun (Lond). 2021;41:1037–48.

    Article  PubMed  Google Scholar 

  3. Lokeshwar SD, Klaassen Z, Saad F. Treatment and trials in non-metastatic castration-resistant prostate cancer. Nat Rev Urol. 2021;18:433–42.

    Article  PubMed  Google Scholar 

  4. Thomsen MK, Busk M. Pre-clinical models to study human prostate Cancer. Cancers (Basel). 2023;15:4212.

  5. Angappulige DH, Mahajan NP, Mahajan K. Epigenetic underpinnings of tumor-immune dynamics in prostate cancer immune suppression. Trends Cancer. 2024;10:369–81.

  6. Zaalberg A, Pottendorfer E, Zwart W, Bergman AM. It takes two to Tango: the interplay between prostate Cancer and its microenvironment from an epigenetic perspective. Cancers (Basel). 2024;16:294.

  7. Feng D, Xiong Q, Wei Q, Yang L. Cellular landscape of tumour microenvironment in prostate cancer. Immunology. 2023;168:199–202.

    Article  PubMed  CAS  Google Scholar 

  8. Kang J, La Manna F, Bonollo F, Sampson N, Alberts IL, Mingels C, Afshar-Oromieh A, Thalmann GN, Karkampouna S. Tumor microenvironment mechanisms and bone metastatic disease progression of prostate cancer. Cancer Lett. 2022;530:156–69.

    Article  PubMed  CAS  Google Scholar 

  9. Li MO, Wolf N, Raulet DH, Akkari L, Pittet MJ, Rodriguez PC, Kaplan RN, Munitz A, Zhang Z, Cheng S, Bhardwaj N. Innate immune cells in the tumor microenvironment. Cancer Cell. 2021;39:725–9.

    Article  PubMed  CAS  Google Scholar 

  10. Lin Q, Choyke PL, Sato N. Visualizing vasculature and its response to therapy in the tumor microenvironment. Theranostics. 2023;13:5223–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Helms E, Onate MK, Sherman MH. Fibroblast heterogeneity in the pancreatic Tumor Microenvironment. Cancer Discov. 2020;10:648–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Piperigkou Z, Karamanos NK. Dynamic interplay between miRNAs and the Extracellular Matrix influences the Tumor Microenvironment. Trends Biochem Sci. 2019;44:1076–88.

    Article  PubMed  CAS  Google Scholar 

  13. Shiao SL, Chu GC, Chung LW. Regulation of prostate cancer progression by the tumor microenvironment. Cancer Lett. 2016;380:340–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Abdul Sater H, Marte JL, Donahue RN, Walter-Rodriguez B, Heery CR, Steinberg SM, Cordes LM, Chun G, Karzai F, Bilusic M et al. Neoadjuvant PROSTVAC prior to radical prostatectomy enhances T-cell infiltration into the tumor immune microenvironment in men with prostate cancer. J Immunother Cancer. 2020;8:e000655.

  15. Wang M, Wisniewski CA, Xiong C, Chhoy P, Goel HL, Kumar A, Zhu LJ, Li R, St Louis PA, Ferreira LM, et al. Therapeutic blocking of VEGF binding to neuropilin-2 diminishes PD-L1 expression to activate antitumor immunity in prostate cancer. Sci Transl Med. 2023;15:eade5855.

    Article  PubMed  CAS  Google Scholar 

  16. Zhang Z, Karthaus WR, Lee YS, Gao VR, Wu C, Russo JW, Liu M, Mota JM, Abida W, Linton E, et al. Tumor Microenvironment-Derived NRG1 promotes Antiandrogen Resistance in prostate Cancer. Cancer Cell. 2020;38:279–e296279.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Javier-DesLoges J, McKay RR, Swafford AD, Sepich-Poore GD, Knight R, Parsons JK. The microbiome and prostate cancer. Prostate Cancer Prostatic Dis. 2022;25:159–64.

    Article  PubMed  Google Scholar 

  18. Hirz T, Mei S, Sarkar H, Kfoury Y, Wu S, Verhoeven BM, Subtelny AO, Zlatev DV, Wszolek MW, Salari K, et al. Dissecting the immune suppressive human prostate tumor microenvironment via integrated single-cell and spatial transcriptomic analyses. Nat Commun. 2023;14:663.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Gu CY, Dai B, Zhu Y, Lin GW, Wang HK, Ye DW, Qin XJ. The novel transcriptomic signature of angiogenesis predicts clinical outcome, tumor microenvironment and treatment response for prostate adenocarcinoma. Mol Med. 2022;28:78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Gevaert T, Van Eycke YR, Vanden Broeck T, Van Poppel H, Salmon I, Rorive S, Muilwijk T, Claessens F, De Ridder D, Joniau S, Decaestecker C. The potential of tumour microenvironment markers to stratify the risk of recurrence in prostate cancer patients. PLoS ONE. 2020;15:e0244663.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Liu Z, Zhong J, Zeng J, Duan X, Lu J, Sun X, Liu Q, Liang Y, Lin Z, Zhong W, et al. Characterization of the m6A-Associated Tumor Immune Microenvironment in prostate Cancer to Aid Immunotherapy. Front Immunol. 2021;12:735170.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Wu T, Wang W, Shi G, Hao M, Wang Y, Yao M, Huang Y, Du L, Zhang X, Ye D, et al. Targeting HIC1/TGF-beta axis-shaped prostate cancer microenvironment restrains its progression. Cell Death Dis. 2022;13:624.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Mizuno K, Beltran H. Future directions for precision oncology in prostate cancer. Prostate. 2022;82(Suppl 1):S86–96.

    PubMed  PubMed Central  CAS  Google Scholar 

  24. Goel HL, Pursell B, Shultz LD, Greiner DL, Brekken RA, Vander Kooi CW, Mercurio AM. P-Rex1 promotes resistance to VEGF/VEGFR-Targeted therapy in prostate Cancer. Cell Rep. 2016;14:2193–208.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Powles T, Yuen KC, Gillessen S, Kadel EE 3rd, Rathkopf D, Matsubara N, Drake CG, Fizazi K, Piulats JM, Wysocki PJ, et al. Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: a randomized phase 3 trial. Nat Med. 2022;28:144–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Agarwal N, McGregor B, Maughan BL, Dorff TB, Kelly W, Fang B, McKay RR, Singh P, Pagliaro L, Dreicer R, et al. Cabozantinib in combination with atezolizumab in patients with metastatic castration-resistant prostate cancer: results from an expansion cohort of a multicentre, open-label, phase 1b trial (COSMIC-021). Lancet Oncol. 2022;23:899–909.

    Article  PubMed  CAS  Google Scholar 

  27. Aparicio AM, Harzstark AL, Corn PG, Wen S, Araujo JC, Tu SM, Pagliaro LC, Kim J, Millikan RE, Ryan C, et al. Platinum-based chemotherapy for variant castrate-resistant prostate cancer. Clin Cancer Res. 2013;19:3621–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Pereira M, Vale N. Repurposing alone and in combination of the antiviral saquinavir with 5-Fluorouracil in prostate and lung cancer cells. Int J Mol Sci. 2022;23:12240.

  29. Sun L, Tuo Z, Chen X, Wang H, Lyu Z, Li G. Identification of cell differentiation trajectory-related gene signature to reveal the prognostic significance and immune landscape in prostate cancer based on multiomics analysis. Heliyon. 2024;10:e27628.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Chen Z, Yang X, Chen Z, Li M, Wang W, Yang R, Wang Z, Ma Y, Xu Y, Ao S, et al. A new histone deacetylase inhibitor remodels the tumor microenvironment by deletion of polymorphonuclear myeloid-derived suppressor cells and sensitizes prostate cancer to immunotherapy. BMC Med. 2023;21:402.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Di Donato M, Zamagni A, Galasso G, Di Zazzo E, Giovannelli P, Barone MV, Zanoni M, Gunelli R, Costantini M, Auricchio F, et al. The androgen receptor/filamin A complex as a target in prostate cancer microenvironment. Cell Death Dis. 2021;12:127.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zhang Y, Zhu S, Du Y, Xu F, Sun W, Xu Z, Wang X, Qian P, Zhang Q, Feng J, Xu Y. RelB upregulates PD-L1 and exacerbates prostate cancer immune evasion. J Exp Clin Cancer Res. 2022;41:66.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Li N, Liu Q, Han Y, Pei S, Cheng B, Xu J, Miao X, Pan Q, Wang H, Guo J, et al. ARID1A loss induces polymorphonuclear myeloid-derived suppressor cell chemotaxis and promotes prostate cancer progression. Nat Commun. 2022;13:7281.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Calcinotto A, Spataro C, Zagato E, Di Mitri D, Gil V, Crespo M, De Bernardis G, Losa M, Mirenda M, Pasquini E, et al. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature. 2018;559:363–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Goulielmaki M, Stokidis S, Anagnostou T, Voutsas IF, Gritzapis AD, Baxevanis CN, Fortis SP. Frequencies of an immunogenic HER-2/neu Epitope of CD8 + T lymphocytes predict favorable clinical outcomes in prostate Cancer. Int J Mol Sci. 2023;24:5954.

  36. Doonan BP, Haque A. HLA Class II Antigen presentation in prostate Cancer cells: a Novel Approach to prostate Tumor Immunotherapy. Open Cancer Immunol J. 2010;3:1–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Foro P, Algara M, Lozano J, Rodriguez N, Sanz X, Torres E, Carles J, Reig A, Membrive I, Quera J, et al. Relationship between radiation-induced apoptosis of T lymphocytes and chronic toxicity in patients with prostate cancer treated by radiation therapy: a prospective study. Int J Radiat Oncol Biol Phys. 2014;88:1057–63.

    Article  PubMed  Google Scholar 

  38. Dorff TB, Narayan V, Forman SJ, Zang PD, Fraietta JA, June CH, Haas NB, Priceman SJ. Novel redirected T-Cell immunotherapies for advanced prostate Cancer. Clin Cancer Res. 2022;28:576–84.

    Article  PubMed  CAS  Google Scholar 

  39. Davidsson S, Ohlson AL, Andersson SO, Fall K, Meisner A, Fiorentino M, Andren O, Rider JR. CD4 helper T cells, CD8 cytotoxic T cells, and FOXP3(+) regulatory T cells with respect to lethal prostate cancer. Mod Pathol. 2013;26:448–55.

    Article  PubMed  CAS  Google Scholar 

  40. Wang H, Mendez L, Morton G, Loblaw A, Chung HT, Cheung P, Mesci A, Escueta V, Petchiny TN, Huang X, et al. Brachytherapy for high grade prostate cancer induces distinct changes in circulating CD4 and CD8 T cells - implications for systemic control. Radiother Oncol. 2024;191:110077.

    Article  PubMed  CAS  Google Scholar 

  41. Cerqueira MA, Ferrari KL, de Mattos AC, Monti CR, Reis LO. T cells CD4+/CD8 + local immune modulation by prostate cancer hemi-cryoablation. World J Urol. 2020;38:673–80.

    Article  PubMed  CAS  Google Scholar 

  42. Shackleton EG, Ali HY, Khan M, Pockley GA, McArdle SE. Novel combinatorial approaches to tackle the immunosuppressive microenvironment of prostate cancer. Cancers (Basel). 2021;13:1145.

  43. Andersen LB, Norgaard M, Rasmussen M, Fredsoe J, Borre M, Ulhoi BP, Sorensen KD. Immune cell analyses of the tumor microenvironment in prostate cancer highlight infiltrating regulatory T cells and macrophages as adverse prognostic factors. J Pathol. 2021;255:155–65.

    Article  PubMed  CAS  Google Scholar 

  44. Ge Q, Zhao Z, Li X, Yang F, Zhang M, Hao Z, Liang C, Meng J. Deciphering the suppressive immune microenvironment of prostate cancer based on CD4 + regulatory T cells: implications for prognosis and therapy prediction. Clin Transl Med. 2024;14:e1552.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Chang M, He Y, Liu C, Lin R, Huang X, Liang D, Zhang J, Lu Y. Downregulation of SEPTIN5 inhibits prostate cancer progression by increasing CD8(+) T cell infiltration. Int J Biol Sci. 2022;18:6035–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Wu X, Zhu Y, Hu C, Du X, Xue W, Chen Y, Dong L, Pan J. Extracellular vesicles related gene HSPH1 exerts anti-tumor effects in prostate cancer via promoting the stress response of CD8 + T cells. Cell Oncol (Dordr). 2024;47:1059–64.

    Article  PubMed  CAS  Google Scholar 

  47. Zhou X, Zou L, Liao H, Luo J, Yang T, Wu J, Chen W, Wu K, Cen S, Lv D, et al. Abrogation of HnRNP L enhances anti-PD-1 therapy efficacy via diminishing PD-L1 and promoting CD8(+) T cell-mediated ferroptosis in castration-resistant prostate cancer. Acta Pharm Sin B. 2022;12:692–707.

    Article  PubMed  CAS  Google Scholar 

  48. Vardeu A, Davis C, McDonald I, Stahlberg G, Thapa B, Piotrowska K, Marshall MA, Evans T, Wheeler V, Sebastian S, Anderson K. Intravenous administration of viral vectors expressing prostate cancer antigens enhances the magnitude and functionality of CD8 + T cell responses. J Immunother Cancer. 2022;10:e005398.

  49. Bohner P, Chevalier MF, Cesson V, Rodrigues-Dias SC, Dartiguenave F, Burruni R, Tawadros T, Valerio M, Lucca I, Nardelli-Haefliger D, et al. Double positive CD4(+)CD8(+) T cells are enriched in Urological Cancers and Favor T Helper-2 polarization. Front Immunol. 2019;10:622.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Lopez-Bujanda ZA, Chaimowitz MG, Armstrong TD, Foote JB, Emens LA, Drake CG. Robust antigen-specific CD8 T cell tolerance to a model prostate cancer neoantigen. Oncoimmunology. 2020;9:1809926.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Das G, Ptacek J, Havlinova B, Nedvedova J, Barinka C, Novakova Z. Targeting prostate Cancer using bispecific T-Cell engagers against prostate-specific membrane Antigen. ACS Pharmacol Transl Sci. 2023;6:1703–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Dejous C, Krishnan UM. Sensors for diagnosis of prostate cancer: looking beyond the prostate specific antigen. Biosens Bioelectron. 2021;173:112790.

    Article  PubMed  CAS  Google Scholar 

  53. Narain TA, Sooriakumaran P. Beyond prostate specific Antigen: new prostate Cancer Screening options. World J Mens Health. 2022;40:66–73.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Joglekar AV, Leonard MT, Jeppson JD, Swift M, Li G, Wong S, Peng S, Zaretsky JM, Heath JR, Ribas A, et al. T cell antigen discovery via signaling and antigen-presenting bifunctional receptors. Nat Methods. 2019;16:191–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Chen JY, Agrawal S, Yi HP, Vallejo D, Agrawal A, Lee AP. Cell-sized lipid vesicles as Artificial Antigen-presenting cells for Antigen-specific T cell activation. Adv Healthc Mater. 2023;12:e2203163.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Feriz AM, Khosrojerdi A, Lotfollahi M, Shamsaki N, GhasemiGol M, HosseiniGol E, Fereidouni M, Rohban MH, Sebzari AR, Saghafi S, et al. Single-cell RNA sequencing uncovers heterogeneous transcriptional signatures in tumor-infiltrated dendritic cells in prostate cancer. Heliyon. 2023;9:e15694.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Bian X, Wang W, Abudurexiti M, Zhang X, Ma W, Shi G, Du L, Xu M, Wang X, Tan C, et al. Integration analysis of single-cell Multi-omics reveals prostate Cancer heterogeneity. Adv Sci (Weinh). 2024;11:e2305724.

    Article  PubMed  Google Scholar 

  58. Merkley SD, Chock CJ, Yang XO, Harris J, Castillo EF. Modulating T cell responses via Autophagy: the intrinsic Influence Controlling the function of both Antigen-presenting cells and T cells. Front Immunol. 2018;9:2914.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Saudi A, Banday V, Zirakzadeh AA, Selinger M, Forsberg J, Holmbom M, Henriksson J, Walden M, Alamdari F, Aljabery F et al. Immune-activated B cells are dominant in prostate cancer. Cancers (Basel). 2023;15:920.

  60. Linde MH, Fan AC, Kohnke T, Trotman-Grant AC, Gurev SF, Phan P, Zhao F, Haddock NL, Nuno KA, Gars EJ, et al. Reprogramming Cancer into Antigen-presenting cells as a Novel Immunotherapy. Cancer Discov. 2023;13:1164–85.

    Article  PubMed  CAS  Google Scholar 

  61. Wang C, Chen L, Fu D, Liu W, Puri A, Kellis M, Yang J. Antigen presenting cells in cancer immunity and mediation of immune checkpoint blockade. Clin Exp Metastasis. 2024;41:333–49.

  62. Zagorulya M, Duong E, Spranger S. Impact of anatomic site on antigen-presenting cells in cancer. J Immunother Cancer. 2020;8:e001204.

  63. Sarkar C, Goswami S, Basu S, Chakroborty D. Angiogenesis inhibition in prostate Cancer: an update. Cancers (Basel). 2020;12:2382.

  64. Melegh Z, Oltean S. Targeting angiogenesis in prostate Cancer. Int J Mol Sci. 2019;20:2676.

  65. Luo Y, Yang Z, Yu Y, Zhang P. HIF1alpha lactylation enhances KIAA1199 transcription to promote angiogenesis and vasculogenic mimicry in prostate cancer. Int J Biol Macromol. 2022;222:2225–43.

    Article  PubMed  CAS  Google Scholar 

  66. Ioannidou E, Moschetta M, Shah S, Parker JS, Ozturk MA, Pappas-Gogos G, Sheriff M, Rassy E, Boussios S. Angiogenesis and anti-angiogenic treatment in prostate cancer: mechanisms of action and molecular targets. Int J Mol Sci. 2021;22:9926.

  67. Cho HD, Kim JH, Park JK, Hong SM, Kim DH, Seo KI. Kochia scoparia seed extract suppresses VEGF-induced angiogenesis via modulating VEGF receptor 2 and PI3K/AKT/mTOR pathways. Pharm Biol. 2019;57:684–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Ene C, Nicolae I, Ene CD. Angiogenic systemic response to the hypoxic microenvironment in prostate tumorigenesis: a pilot study. Exp Ther Med. 2023;26:483.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Baghaie L, Haxho F, Leroy F, Lewis B, Wawer A, Minhas S, Harless WW, Szewczuk MR. Contemporaneous perioperative inflammatory and angiogenic cytokine profiles of surgical breast, colorectal, and prostate cancer patients: clinical implications. Cells. 2023;12:2767.

  70. Fang L, Li D, Yin J, Pan H, Ye H, Bowman J, Capaldo B, Kelly K. TMPRSS2-ERG promotes the initiation of prostate cancer by suppressing oncogene-induced senescence. Cancer Gene Ther. 2022;29:1463–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Miyake M, Furuya H, Onishi S, Hokutan K, Anai S, Chan O, Shi S, Fujimoto K, Goodison S, Cai W, Rosser CJ. Monoclonal antibody against CXCL1 (HL2401) as a Novel Agent in suppressing IL6 expression and Tumoral Growth. Theranostics. 2019;9:853–67.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Ciummo SL, Sorrentino C, Fieni C, Di Carlo E. Interleukin-30 subverts prostate cancer-endothelium crosstalk by fostering angiogenesis and activating immunoregulatory and oncogenic signaling pathways. J Exp Clin Cancer Res. 2023;42:336.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Bhattacharya IS, Taghavi Azar Sharabiani M, Alonzi R, Hoskin PJ. Hypoxia and angiogenic biomarkers in prostate cancer after external beam radiotherapy (EBRT) alone or combined with high-dose-rate brachytherapy boost (HDR-BTb). Radiother Oncol. 2019;137:38–44.

    Article  PubMed  CAS  Google Scholar 

  74. Rios-Colon L, Kumar P, Kim S, Sharma M, Su Y, Kumar A, Singh S, Stocks N, Liu L, Joshi M et al. Carnitine palmitoyltransferase 1 regulates prostate cancer growth under hypoxia. Cancers (Basel). 2021;13:6302.

  75. Xu W, Qian J, Zeng F, Li S, Guo W, Chen L, Li G, Zhang Z, Wang QJ, Deng F. Protein kinase ds promote tumor angiogenesis through mast cell recruitment and expression of angiogenic factors in prostate cancer microenvironment. J Exp Clin Cancer Res. 2019;38:114.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Wang H, Li N, Liu Q, Guo J, Pan Q, Cheng B, Xu J, Dong B, Yang G, Yang B, et al. Antiandrogen treatment induces stromal cell reprogramming to promote castration resistance in prostate cancer. Cancer Cell. 2023;41:1345–e13621349.

    Article  PubMed  CAS  Google Scholar 

  77. Nilsson EM, Laursen KB, Whitchurch J, McWilliam A, Odum N, Persson JL, Heery DM, Gudas LJ, Mongan NP. MiR137 is an androgen regulated repressor of an extended network of transcriptional coregulators. Oncotarget. 2015;6:35710–25.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Panus A, Simionescu CE, Dragoescu PO, Stepan AE. VEGF immunoexpression in prostate adenocarcinoma. Curr Health Sci J. 2021;47:89–95.

    PubMed  PubMed Central  Google Scholar 

  79. Drobkova H, Jurecekova J, Sivonova MK, Mazuchova J, Skorvanova M, Sarlinova M, Halasova E, Kliment J. Associations between Gene polymorphisms of vascular endothelial growth factor and prostate Cancer. Anticancer Res. 2019;39:2903–9.

    Article  PubMed  CAS  Google Scholar 

  80. Catena R, Larzabal L, Larrayoz M, Molina E, Hermida J, Agorreta J, Montes R, Pio R, Montuenga LM, Calvo A. VEGF(1)(2)(1)b and VEGF(1)(6)(5)b are weakly angiogenic isoforms of VEGF-A. Mol Cancer. 2010;9:320.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Huang TF, Wang SW, Lai YW, Liu SC, Chen YJ, Hsueh TY, Lin CC, Lin CH, Chung CH. 4-Acetylantroquinonol B suppresses prostate cancer growth and angiogenesis via a VEGF/PI3K/ERK/mTOR-Dependent signaling pathway in subcutaneous xenograft and in vivo angiogenesis models. Int J Mol Sci. 2022;23:1446.

  82. Basagiannis D, Zografou S, Murphy C, Fotsis T, Morbidelli L, Ziche M, Bleck C, Mercer J, Christoforidis S. VEGF induces signalling and angiogenesis by directing VEGFR2 internalisation through macropinocytosis. J Cell Sci. 2016;129:4091–104.

    Article  PubMed  CAS  Google Scholar 

  83. Larsson P, Syed Khaja AS, Semenas J, Wang T, Sarwar M, Dizeyi N, Simoulis A, Hedblom A, Wai SN, Odum N, Persson JL. The functional interlink between AR and MMP9/VEGF signaling axis is mediated through PIP5K1alpha/pAKT in prostate cancer. Int J Cancer. 2020;146:1686–99.

    Article  PubMed  CAS  Google Scholar 

  84. Baci D, Bruno A, Cascini C, Gallazzi M, Mortara L, Sessa F, Pelosi G, Albini A, Noonan DM. Acetyl-L-Carnitine downregulates invasion (CXCR4/CXCL12, MMP-9) and angiogenesis (VEGF, CXCL8) pathways in prostate cancer cells: rationale for prevention and interception strategies. J Exp Clin Cancer Res. 2019;38:464.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Trujillo-Rojas L, Fernandez-Novell JM, Blanco-Prieto O, Rigau T, Del Rivera MM, Rodriguez-Gil JE. The onset of age-related benign prostatic hyperplasia is concomitant with increased serum and prostatic expression of VEGF in rats: potential role of VEGF as a marker for early prostatic alterations. Theriogenology. 2022;183:69–78.

    Article  PubMed  CAS  Google Scholar 

  86. Giacomini A, Grillo E, Rezzola S, Ribatti D, Rusnati M, Ronca R, Presta M. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev. 2021;101:569–610.

    Article  PubMed  CAS  Google Scholar 

  87. Labanca E, Yang J, Shepherd PDA, Wan X, Starbuck MW, Guerra LD, Anselmino N, Bizzotto JA, Dong J, Chinnaiyan AM, et al. Fibroblast growth factor receptor 1 drives the metastatic progression of prostate Cancer. Eur Urol Oncol. 2022;5:164–75.

    Article  PubMed  Google Scholar 

  88. Ferguson HR, Smith MP, Francavilla C. Fibroblast growth factor receptors (FGFRs) and Noncanonical partners in cancer signaling. Cells. 2021;10:1201.

  89. Teishima J, Hayashi T, Nagamatsu H, Shoji K, Shikuma H, Yamanaka R, Sekino Y, Goto K, Inoue S, Matsubara A. Fibroblast growth factor family in the progression of prostate cancer. J Clin Med. 2019;8:183.

  90. Yu L, Toriseva M, Afshan S, Cangiano M, Fey V, Erickson A, Seikkula H, Alanen K, Taimen P, Ettala O et al. Increased expression and altered cellular localization of fibroblast growth factor receptor-like 1 (FGFRL1) are Associated with prostate cancer progression. Cancers (Basel). 2022;14:278.

  91. Ghinea N, Robin B, Pichon C, Leclere R, Nicolas A, Chnecker C, Cote JF, Guillonneau B, Radu A. Vasa Nervorum angiogenesis in prostate cancer with perineural invasion. Prostate. 2019;79:640–6.

    Article  PubMed  CAS  Google Scholar 

  92. Wang C, Liu Z, Ke Y, Wang F. Intrinsic FGFR2 and ectopic FGFR1 signaling in the prostate and prostate Cancer. Front Genet. 2019;10:12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Stangis MM, Colah AN, McLean DT, Halberg RB, Collier LS, Ricke WA. Potential roles of FGF5 as a candidate therapeutic target in prostate cancer. Am J Clin Exp Urol. 2023;11:452–66.

    PubMed  PubMed Central  Google Scholar 

  94. Kregel S, Kiriluk KJ, Rosen AM, Cai Y, Reyes EE, Otto KB, Tom W, Paner GP, Szmulewitz RZ. Vander Griend DJ: Sox2 is an androgen receptor-repressed gene that promotes castration-resistant prostate cancer. PLoS ONE. 2013;8:e53701.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Cheng J, Ye H, Liu Z, Xu C, Zhang Z, Liu Y, Sun Y. Platelet-derived growth factor-BB accelerates prostate cancer growth by promoting the proliferation of mesenchymal stem cells. J Cell Biochem. 2013;114:1510–8.

    Article  PubMed  CAS  Google Scholar 

  96. Iqbal S, Zhang S, Driss A, Liu ZR, Kim HR, Wang Y, Ritenour C, Zhau HE, Kucuk O, Chung LW, Wu D. PDGF upregulates Mcl-1 through activation of beta-catenin and HIF-1alpha-dependent signaling in human prostate cancer cells. PLoS ONE. 2012;7:e30764.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Rosenberg A, Mathew P. Imatinib and prostate cancer: lessons learned from targeting the platelet-derived growth factor receptor. Expert Opin Investig Drugs. 2013;22:787–94.

    Article  PubMed  CAS  Google Scholar 

  98. Russell MR, Liu Q, Fatatis A. Targeting the alpha receptor for platelet-derived growth factor as a primary or combination therapy in a preclinical model of prostate cancer skeletal metastasis. Clin Cancer Res. 2010;16:5002–10.

    Article  PubMed  CAS  Google Scholar 

  99. Najy AJ, Won JJ, Movilla LS, Kim HR. Differential tumorigenic potential and matriptase activation between PDGF B versus PDGF D in prostate cancer. Mol Cancer Res. 2012;10:1087–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P. Endothelial cell metabolism. Physiol Rev. 2018;98:3–58.

    Article  PubMed  CAS  Google Scholar 

  101. Bhowmick S, Bhowmick NA. RARgamma: the bone of contention for endothelial cells in prostate Cancer metastasis. Cancer Res. 2022;82:2975–6.

    Article  PubMed  CAS  Google Scholar 

  102. Zhao R, Bei X, Yang B, Wang X, Jiang C, Shi F, Wang X, Zhu Y, Jing Y, Han B, et al. Endothelial cells promote metastasis of prostate cancer by enhancing autophagy. J Exp Clin Cancer Res. 2018;37:221.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Juan-Rivera MC, Martinez-Ferrer M. Integrin inhibitors in prostate cancer. Cancers (Basel). 2018;10:44.

  104. Klein D, Schmitz T, Verhelst V, Panic A, Schenck M, Reis H, Drab M, Sak A, Herskind C, Maier P, Jendrossek V. Endothelial Caveolin-1 regulates the radiation response of epithelial prostate tumors. Oncogenesis. 2015;4:e148.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Ji S, Wu W, Jiang Q. Crosstalk between endothelial cells and Tumor cells: a new era in prostate cancer progression. Int J Mol Sci. 2023;24:16893.

  106. Fu S, Liu T, Lv C, Fu C, Zeng R, Kakehi Y, Kulkarni P, Getzenberg RH, Zeng Y. Stromal-epithelial interactions in prostate cancer: overexpression of PAGE4 in stromal cells inhibits the invasive ability of epithelial cells. J Cell Biochem. 2020;121:4406–18.

    Article  PubMed  CAS  Google Scholar 

  107. Enriquez C, Cancila V, Ferri R, Sulsenti R, Fischetti I, Milani M, Ostano P, Gregnanin I, Mello-Grand M, Berrino E, et al. Castration-Induced downregulation of SPARC in stromal cells drives neuroendocrine differentiation of prostate Cancer. Cancer Res. 2021;81:4257–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Tang Q, Cheng B, Dai R, Wang R. The role of androgen receptor in Cross Talk between stromal cells and prostate Cancer epithelial cells. Front Cell Dev Biol. 2021;9:729498.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Frankenstein Z, Basanta D, Franco OE, Gao Y, Javier RA, Strand DW, Lee M, Hayward SW, Ayala G, Anderson ARA. Stromal reactivity differentially drives tumour cell evolution and prostate cancer progression. Nat Ecol Evol. 2020;4:870–84.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Chen L, Yang F, Li T, Xiao P, Han ZJ, Shu LF, Yuan ZZ, Liu WJ, Long YQ. Extracellular histone promotes prostate Cancer Migration and epithelial-mesenchymal transition through NF-kappaB-mediated inflammatory responses. Chemotherapy. 2019;64:177–86.

    Article  PubMed  CAS  Google Scholar 

  111. Orme JJ, Huang H. Microenvironment-mediated resistance to Anti-androgen Therapy. Cancer Cell. 2020;38:155–7.

    Article  PubMed  CAS  Google Scholar 

  112. Xu H, Sun Y, You B, Huang CP, Ye D, Chang C. Androgen receptor reverses the oncometabolite R-2-hydroxyglutarate-induced prostate cancer cell invasion via suppressing the circRNA-51217/miRNA-646/TGFbeta1/p-Smad2/3 signaling. Cancer Lett. 2020;472:151–64.

    Article  PubMed  CAS  Google Scholar 

  113. Pederzoli F, Raffo M, Pakula H, Ravera F, Nuzzo PV, Loda M. Stromal cells in prostate cancer pathobiology: friends or foes? Br J Cancer. 2023;128:930–9.

    Article  PubMed  Google Scholar 

  114. Leach DA, Buchanan G. Protocols for studies on stromal cells in prostate Cancer. Methods Mol Biol. 2018;1786:207–18.

    Article  PubMed  CAS  Google Scholar 

  115. ChallaSivaKanaka S, Vickman RE, Kakarla M, Hayward SW, Franco OE. Fibroblast heterogeneity in prostate carcinogenesis. Cancer Lett. 2022;525:76–83.

    Article  PubMed  CAS  Google Scholar 

  116. Vitale G, Caraglia M, Jung V, Kamradt J, Gentilini D, Di Martino MT, Dicitore A, Abate M, Tagliaferri P, Itro A et al. Molecular characterization of cancer associated fibroblasts in prostate cancer. Cancers (Basel). 2022;14:2943.

  117. Shen T, Li Y, Zhu S, Yu J, Zhang B, Chen X, Zhang Z, Ma Y, Niu Y, Shang Z. YAP1 plays a key role of the conversion of normal fibroblasts into cancer-associated fibroblasts that contribute to prostate cancer progression. J Exp Clin Cancer Res. 2020;39:36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Liu X, Tang J, Peng L, Nie H, Zhang Y, Liu P. Cancer-associated fibroblasts promote malignant phenotypes of prostate cancer cells via autophagy: Cancer-associated fibroblasts promote prostate cancer development. Apoptosis. 2023;28:881–91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Fontana F, Anselmi M, Limonta P. Adipocytes reprogram prostate cancer stem cell machinery. J Cell Commun Signal. 2023;17:915–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Gwak J, Jeong H, Lee K, Shin JY, Sim T, Na J, Kim J, Ju BG. SFMBT2-Mediated infiltration of preadipocytes and TAMs in prostate cancer. Cancers (Basel). 2020;12:2718.

  121. Laurent V, Guerard A, Mazerolles C, Le Gonidec S, Toulet A, Nieto L, Zaidi F, Majed B, Garandeau D, Socrier Y, et al. Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat Commun. 2016;7:10230.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Laurent V, Toulet A, Attane C, Milhas D, Dauvillier S, Zaidi F, Clement E, Cinato M, Le Gonidec S, Guerard A, et al. Periprostatic adipose tissue favors prostate Cancer Cell Invasion in an obesity-dependent manner: role of oxidative stress. Mol Cancer Res. 2019;17:821–35.

    Article  PubMed  CAS  Google Scholar 

  123. Penet MF, Kakkad S, Pathak AP, Krishnamachary B, Mironchik Y, Raman V, Solaiyappan M, Bhujwalla ZM. Structure and function of a prostate Cancer dissemination-permissive Extracellular Matrix. Clin Cancer Res. 2017;23:2245–54.

    Article  PubMed  CAS  Google Scholar 

  124. Luthold C, Hallal T, Labbe DP, Bordeleau F. The extracellular matrix stiffening: a trigger of prostate cancer progression and castration resistance? Cancers (Basel). 2022;14:2887.

  125. Pang X, Xie R, Zhang Z, Liu Q, Wu S, Cui Y. Identification of SPP1 as an Extracellular Matrix signature for metastatic castration-resistant prostate Cancer. Front Oncol. 2019;9:924.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Xinzhou H, Ning Y, Ou W, Xiaodan L, Fumin Y, Huitu L, Wei Z. RKIp inhibits the migration and invasion of human prostate cancer PC-3 M cells through regulation of extracellular matrix. Mol Biol (Mosk). 2011;45:1004–11.

    Article  PubMed  CAS  Google Scholar 

  127. Salachan PV, Sorensen KD. Dysbiotic microbes and how to find them: a review of microbiome profiling in prostate cancer. J Exp Clin Cancer Res. 2022;41:31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Yow MA, Tabrizi SN, Severi G, Bolton DM, Pedersen J, Australian Prostate Cancer B, Giles GG, Southey MC. Characterisation of microbial communities within aggressive prostate cancer tissues. Infect Agent Cancer. 2017;12:4.

    Article  PubMed  PubMed Central  Google Scholar 

  129. McCulloch JA, Trinchieri G. Gut bacteria enable prostate cancer growth. Science. 2021;374:154–5.

    Article  PubMed  CAS  Google Scholar 

  130. Pernigoni N, Zagato E, Calcinotto A, Troiani M, Mestre RP, Cali B, Attanasio G, Troisi J, Minini M, Mosole S, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374:216–24.

  131. Lee J, Wickes BL, Fu J, Brockman NE, Garg H, Jobin C, Johson-Pais T, Leach R, Lai Z, Liss MA. Prevalence of genotoxic bacteria in men undergoing biopsy for prostate cancer. Prostate. 2023;83:663–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Bruggemann H, Al-Zeer MA. Bacterial signatures and their inflammatory potentials associated with prostate cancer. APMIS. 2020;128:80–91.

    Article  PubMed  Google Scholar 

  133. Altwaijry N, Somani S, Dufes C. Targeted nonviral gene therapy in prostate cancer. Int J Nanomed. 2018;13:5753–67.

    Article  CAS  Google Scholar 

  134. Khatami A, Nahand JS, Kiani SJ, Khoshmirsafa M, Moghoofei M, Khanaliha K, Tavakoli A, Emtiazi N, Bokharaei-Salim F. Human papilloma virus (HPV) and prostate cancer (PCa): the potential role of HPV gene expression and selected cellular MiRNAs in PCa development. Microb Pathog. 2022;166:105503.

    Article  PubMed  CAS  Google Scholar 

  135. Mygatt JG, Singhal A, Sukumar G, Dalgard CL, Kaleeba JA. Oncogenic herpesvirus HHV-8 promotes androgen-independent prostate cancer growth. Cancer Res. 2013;73:5695–708.

    Article  PubMed  CAS  Google Scholar 

  136. Jenkins FJ, Minas TZ, Tang W, Dorsey TH, Ambs S. Human herpesvirus 8 infection is associated with prostate cancer among IFNL4-DeltaG carriers. Prostate Cancer Prostatic Dis. 2023;26:338–46.

    Article  PubMed  CAS  Google Scholar 

  137. Sfanos KS, Yegnasubramanian S, Nelson WG, De Marzo AM. The inflammatory microenvironment and microbiome in prostate cancer development. Nat Rev Urol. 2018;15:11–24.

    Article  PubMed  Google Scholar 

  138. Che B, Zhang W, Xu S, Yin J, He J, Huang T, Li W, Yu Y, Tang K. Prostate microbiota and prostate Cancer: a New Trend in Treatment. Front Oncol. 2021;11:805459.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Yu H, Meng H, Zhou F, Ni X, Shen S, Das UN. Urinary microbiota in patients with prostate cancer and benign prostatic hyperplasia. Arch Med Sci. 2015;11:385–94.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Severi G, Shannon BA, Hoang HN, Baglietto L, English DR, Hopper JL, Pedersen J, Southey MC, Sinclair R, Cohen RJ, Giles GG. Plasma concentration of Propionibacterium acnes antibodies and prostate cancer risk: results from an Australian population-based case-control study. Br J Cancer. 2010;103:411–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Davidsson S, Molling P, Rider JR, Unemo M, Karlsson MG, Carlsson J, Andersson SO, Elgh F, Soderquis B, Andren O. Frequency and typing of Propionibacterium acnes in prostate tissue obtained from men with and without prostate cancer. Infect Agent Cancer. 2016;11:26.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Esteves AM, Papaevangelou E, Smolarek D, Dasgupta P, Galustian C. Cytotopic (Cyto-) IL-15 as a new immunotherapy for prostate Cancer: recombinant production in Escherichia coli and Purification. Front Mol Biosci. 2021;8:755764.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Cao Z, Liu J. Bacteria and bacterial derivatives as drug carriers for cancer therapy. J Control Release. 2020;326:396–407.

    Article  PubMed  CAS  Google Scholar 

  144. Liu Z, Ma Y, Ye J, Li G, Kang X, Xie W, Wang X. Drug delivery systems for enhanced tumour treatment by eliminating intra-tumoral bacteria. J Mater Chem B. 2024;12:1194–207.

    Article  PubMed  CAS  Google Scholar 

  145. Howard FHN, Al-Janabi H, Patel P, Cox K, Smith E, Vadakekolathu J, Pockley AG, Conner J, Nohl JF, Allwood DA, et al. Nanobugs as drugs: bacterial derived nanomagnets enhance Tumor Targeting and Oncolytic Activity of HSV-1 Virus. Small. 2022;18:e2104763.

    Article  PubMed  Google Scholar 

  146. Liu Y, Yang C, Zhang Z, Jiang H. Gut microbiota dysbiosis accelerates prostate Cancer progression through increased LPCAT1 expression and enhanced DNA repair pathways. Front Oncol. 2021;11:679712.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Wang G, Liu Y, Liu S, Lin Y, Hu C. Oncolyic virotherapy for prostate cancer: lighting a fire in winter. Int J Mol Sci. 2022;23:12647.

  148. Marcus JL, Chao CR, Leyden WA, Xu L, Klein DB, Horberg MA, Towner WJ, Quesenberry CP Jr., Abrams DI, Van Den Eeden SK, Silverberg MJ. Prostate cancer incidence and prostate-specific antigen testing among HIV-positive and HIV-negative men. J Acquir Immune Defic Syndr. 2014;66:495–502.

    Article  PubMed  Google Scholar 

  149. Sadri Nahand J, Esghaei M, Hamidreza Monavari S, Moghoofei M, Jalal Kiani S, Mostafaei S, Mirzaei H, Bokharaei-Salim F. The assessment of a possible link between HPV-mediated inflammation, apoptosis, and angiogenesis in prostate cancer. Int Immunopharmacol. 2020;88:106913.

    Article  PubMed  CAS  Google Scholar 

  150. Murphy AB, Bhatia R, Martin IK, Klein DA, Hollowell CMP, Nyame Y, Dielubanza E, Achenbach C, Kittles RA. Are HIV-infected men vulnerable to prostate cancer treatment disparities? Cancer Epidemiol Biomarkers Prev. 2014;23:2009–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Opeyemi Bello R, Willis-Powell L, James O, Sharma A, Marsh E, Ellis L, Gaston K, Siddiqui Y. Does human papillomavirus play a causative role in prostate cancer? a systematic review using bradford hill’s criteria. Cancers (Basel). 2023;15:3897.

  152. Sun J, Xiang J, An Y, Xu J, Xiong Y, Wang S, Xia Q. Unveiling the Association between HPV and Pan-cancers: a bidirectional two-sample mendelian randomization study. Cancers (Basel). 2023;15:5147.

  153. Kypta RM, Waxman J. Wnt/β-catenin signalling in prostate cancer. Nat Rev Urol. 2012;9:418–28.

    Article  PubMed  CAS  Google Scholar 

  154. Kaplan Z, Zielske SP, Ibrahim KG, Cackowski FC. Wnt and beta-catenin signaling in the bone metastasis of prostate Cancer. Life (Basel). 2021;11:1099.

  155. Ma F, Arai S, Wang K, Calagua C, Yuan AR, Poluben L, Gu Z, Russo JW, Einstein DJ, Ye H, et al. Autocrine canonical wnt signaling primes noncanonical signaling through ROR1 in metastatic castration-resistant prostate cancer. Cancer Res. 2022;82:1518–33.

  156. Koushyar S, Meniel VS, Phesse TJ, Pearson HB. Exploring the wnt pathway as a therapeutic target for prostate cancer. Biomolecules. 2022;12:309.

  157. Roudsari NM, Lashgari NA, Momtaz S, Abaft S, Jamali F, Safaiepour P, Narimisa K, Jackson G, Bishayee A, Rezaei N et al. Inhibitors of the PI3K/Akt/mTOR pathway in prostate cancer chemoprevention and intervention. Pharmaceutics. 2021;13:1195.

  158. Chen H, Zhou L, Wu X, Li R, Wen J, Sha J, Wen X. The PI3K/AKT pathway in the pathogenesis of prostate cancer. Front Biosci (Landmark Ed). 2016;21:1084–91.

    Article  PubMed  CAS  Google Scholar 

  159. Chaves LP, Melo CM, Saggioro FP, Reis RBD, Squire JA. Epithelial-mesenchymal transition signaling and prostate cancer stem cells: emerging biomarkers and opportunities for precision therapeutics. Genes (Basel). 2021;12:1900.

  160. Schneider JA, Logan SK. Revisiting the role of Wnt/beta-catenin signaling in prostate cancer. Mol Cell Endocrinol. 2018;462:3–8.

    Article  PubMed  CAS  Google Scholar 

  161. Ding L, Lin Y, Chen X, Wang R, Lu H, Wang H, Luo W, Lu Z, Xia L, Zhou X, et al. circPHF16 suppresses prostate cancer metastasis via modulating miR-581/RNF128/Wnt/beta-catenin pathway. Cell Signal. 2023;102:110557.

    Article  PubMed  CAS  Google Scholar 

  162. Jianfeng W, Yutao W, Jianbin B. TACR2 is associated with the immune microenvironment and inhibits migration and proliferation via the Wnt/beta-catenin signaling pathway in prostate cancer. Cancer Cell Int. 2021;21:415.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Jianfeng W, Yutao W, Jianbin B. TACR2 is associated with the immune microenvironment and inhibits migration and proliferation via the Wnt/β-catenin signaling pathway in prostate cancer. Cancer Cell Int. 2021;21:415.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Jiang Z, Zhang Y, Chen X, Wu P, Chen D. Inactivation of the Wnt/beta-catenin signaling pathway underlies inhibitory role of microRNA-129-5p in epithelial-mesenchymal transition and angiogenesis of prostate cancer by targeting ZIC2. Cancer Cell Int. 2019;19:271.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Chatterjee A, Paul S, Bisht B, Bhattacharya S, Sivasubramaniam S, Paul MK. Advances in targeting the WNT/beta-catenin signaling pathway in cancer. Drug Discov Today. 2022;27:82–101.

    Article  PubMed  CAS  Google Scholar 

  166. Wang Y, Wang J, Tang Q, Ren G. Identification of UBE2C as hub gene in driving prostate cancer by integrated bioinformatics analysis. PLoS ONE. 2021;16:e0247827.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Glaviano A, Foo ASC, Lam HY, Yap KCH, Jacot W, Jones RH, Eng H, Nair MG, Makvandi P, Geoerger B, et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. 2023;22:138.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Yu L, Wei J, Liu P. Attacking the PI3K/Akt/mTOR signaling pathway for targeted therapeutic treatment in human cancer. Semin Cancer Biol. 2022;85:69–94.

    Article  PubMed  CAS  Google Scholar 

  169. Kumar D, Shankar S, Srivastava RK. Rottlerin induces autophagy and apoptosis in prostate cancer stem cells via PI3K/Akt/mTOR signaling pathway. Cancer Lett. 2014;343:179–89.

    Article  PubMed  CAS  Google Scholar 

  170. Tan M, Xu J, Siddiqui J, Feng F, Sun Y. Depletion of SAG/RBX2 E3 ubiquitin ligase suppresses prostate tumorigenesis via inactivation of the PI3K/AKT/mTOR axis. Mol Cancer. 2016;15:81.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Yeh YH, Hsiao HF, Yeh YC, Chen TW, Li TK. Inflammatory interferon activates HIF-1alpha-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway. J Exp Clin Cancer Res. 2018;37:70.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Xu W, Lu M, Xie S, Zhou D, Zhu M, Liang C. Endoplasmic reticulum stress promotes prostate Cancer cells to Release Exosome and Up-regulate PD-L1 expression via PI3K/Akt Signaling Pathway in macrophages. J Cancer. 2023;14:1062–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Ci X, Xing C, Zhang B, Zhang Z, Ni JJ, Zhou W, Dong JT. KLF5 inhibits angiogenesis in PTEN-deficient prostate cancer by attenuating AKT activation and subsequent HIF1α accumulation. Mol Cancer. 2015;14:91.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Pungsrinont T, Kallenbach J, Baniahmad A. Role of PI3K-AKT-mTOR pathway as a pro-survival signaling and resistance-mediating mechanism to therapy of prostate cancer. Int J Mol Sci. 2021;22:11088.

  175. Raith F, O’Donovan DH, Lemos C, Politz O, Haendler B. Addressing the reciprocal crosstalk between the AR and the PI3K/AKT/mTOR signaling pathways for prostate cancer treatment. Int J Mol Sci. 2023;24:2289.

  176. Davis JS, Nastiuk KL, Krolewski JJ. TNF is necessary for castration-induced prostate regression, whereas TRAIL and FasL are dispensable. Mol Endocrinol. 2011;25:611–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Bandil K, Singhal P, Dogra A, Rawal SK, Doval DC, Varshney AK, Bharadwaj M. Association of SNPs/haplotypes in promoter of TNF A and IL-10 gene together with life style factors in prostate cancer progression in Indian population. Inflamm Res. 2017;66:1085–97.

    Article  PubMed  CAS  Google Scholar 

  178. Bakshi S, Zhang X, Godoy-Tundidor S, Cheng RY, Sartor MA, Medvedovic M, Ho SM. Transcriptome analyses in normal prostate epithelial cells exposed to low-dose cadmium: oncogenic and immunomodulations involving the action of tumor necrosis factor. Environ Health Perspect. 2008;116:769–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  179. Liu VWS, Yau WL, Tam CW, Yao KM, Shiu SYW. Melatonin inhibits androgen receptor splice variant-7 (AR-V7)-induced nuclear factor-kappa B (NF-κB) activation and NF-κB activator-induced AR-V7 expression in prostate cancer cells: potential implications for the use of melatonin in castration-resistant prostate cancer (CRPC) therapy. Int J Mol Sci. 2017;18:1130.

  180. Jain G, Cronauer MV, Schrader M, Möller P, Marienfeld RB. NF-κB signaling in prostate cancer: a promising therapeutic target? World J Urol. 2012;30:303–10.

    Article  PubMed  CAS  Google Scholar 

  181. Sethi JK, Hotamisligil GS. Metabolic messengers: tumour necrosis factor. Nat Metab. 2021;3:1302–12.

    Article  PubMed  CAS  Google Scholar 

  182. Romagny S, Bouaouiche S, Lucchi G, Ducoroy P, Bertoldo JB, Terenzi H, Bettaieb A, Plenchette S. S-Nitrosylation of cIAP1 switches Cancer Cell Fate from TNFα/TNFR1-Mediated cell survival to cell death. Cancer Res. 2018;78:1948–57.

    Article  PubMed  CAS  Google Scholar 

  183. Lu L, Tang D, Wang L, Huang LQ, Jiang GS, Xiao XY, Zeng FQ. Gambogic acid inhibits TNF-alpha-induced invasion of human prostate cancer PC3 cells in vitro through PI3K/Akt and NF-kappaB signaling pathways. Acta Pharmacol Sin. 2012;33:531–41.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Giampietri C, Petrungaro S, Padula F, D’Alessio A, Marini ES, Facchiano A, Filippini A, Ziparo E. Autophagy modulators sensitize prostate epithelial cancer cell lines to TNF-alpha-dependent apoptosis. Apoptosis. 2012;17:1210–22.

    Article  PubMed  CAS  Google Scholar 

  185. Cortesi F, Delfanti G, Grilli A, Calcinotto A, Gorini F, Pucci F, Lucianò R, Grioni M, Recchia A, Benigni F, et al. Bimodal CD40/Fas-Dependent crosstalk between iNKT cells and Tumor-Associated macrophages impairs prostate Cancer Progression. Cell Rep. 2018;22:3006–20.

    Article  PubMed  CAS  Google Scholar 

  186. Park IJ, Kim MJ, Park OJ, Park MG, Choe W, Kang I, Kim SS, Ha J. Cryptotanshinone sensitizes DU145 prostate cancer cells to Fas(APO1/CD95)-mediated apoptosis through Bcl-2 and MAPK regulation. Cancer Lett. 2010;298:88–98.

    Article  PubMed  CAS  Google Scholar 

  187. Wellinger LC, Hogg SJ, Newman DM, Friess T, Geiss D, Michie J, Ramsbottom KM, Bacac M, Fauti T, Marbach D, et al. BET inhibition enhances TNF-Mediated Antitumor Immunity. Cancer Immunol Res. 2022;10:87–107.

    Article  PubMed  CAS  Google Scholar 

  188. Elia AR, Grioni M, Basso V, Curnis F, Freschi M, Corti A, Mondino A, Bellone M. Targeting Tumor vasculature with TNF leads effector T cells to the Tumor and enhances therapeutic efficacy of Immune Checkpoint blockers in Combination with adoptive cell therapy. Clin Cancer Res. 2018;24:2171–81.

    Article  PubMed  CAS  Google Scholar 

  189. Griffith TS, Kawakita M, Tian J, Ritchey J, Tartaglia J, Sehgal I, Thompson TC, Zhao W, Ratliff TL. Inhibition of murine prostate tumor growth and activation of immunoregulatory cells with recombinant canarypox viruses. J Natl Cancer Inst. 2001;93:998–1007.

    Article  PubMed  CAS  Google Scholar 

  190. Lai H, Zeng D, Liu C, Zhang Q, Wang X, Chen T. Selenium-containing ruthenium complex synergizes with natural killer cells to enhance immunotherapy against prostate cancer via activating TRAIL/FasL signaling. Biomaterials. 2019;219:119377.

    Article  PubMed  CAS  Google Scholar 

  191. Zhu D, Gao W, Zhang Z. MicroRNA-1180 is associated with growth and apoptosis in prostate cancer via TNF receptor associated factor 1 expression regulation and nuclear factor-κB signaling pathway activation. Oncol Lett. 2018;15:4775–80.

    PubMed  PubMed Central  Google Scholar 

  192. Shankar S, Ganapathy S, Srivastava RK: ‘s Note, editors. Sulforaphane Enhances the Therapeutic Potential of TRAIL in Prostate Cancer Orthotopic Model through Regulation of Apoptosis, Metastasis, and Angiogenesis. Clin Cancer Res 2020, 26:312.

  193. Shankar S, Ganapathy S, Chen Q, Srivastava RK. Curcumin sensitizes TRAIL-resistant xenografts: molecular mechanisms of apoptosis, metastasis and angiogenesis. Mol Cancer. 2008;7:16.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Krolewski JJ, Singh S, Sha K, Jaiswal N, Turowski SG, Pan C, Rich LJ, Seshadri M, Nastiuk KL. TNF signaling is required for Castration-Induced vascular damage preceding prostate cancer regression. Cancers (Basel). 2022;14:6020.

  195. Mu HQ, He YH, Wang SB, Yang S, Wang YJ, Nan CJ, Bao YF, Xie QP, Chen YH. MiR-130b/TNF-alpha/NF-kappaB/VEGFA loop inhibits prostate cancer angiogenesis. Clin Transl Oncol. 2020;22:111–21.

    Article  PubMed  CAS  Google Scholar 

  196. He MX, Cuoco MS, Crowdis J, Bosma-Moody A, Zhang Z, Bi K, Kanodia A, Su MJ, Ku SY, Garcia MM, et al. Transcriptional mediators of treatment resistance in lethal prostate cancer. Nat Med. 2021;27:426–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  197. Hankey W, Chen Z, Wang Q. Shaping chromatin states in prostate Cancer by Pioneer transcription factors. Cancer Res. 2020;80:2427–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Staal J, Beyaert R. Inflammation and NF-kappaB signaling in prostate Cancer: mechanisms and clinical implications. Cells. 2018;7:122.

  199. Thomas-Jardin SE, Dahl H, Nawas AF, Bautista M, Delk NA. NF-kappaB signaling promotes castration-resistant prostate cancer initiation and progression. Pharmacol Ther. 2020;211:107538.

    Article  PubMed  CAS  Google Scholar 

  200. Zhong W, Wu K, Long Z, Zhou X, Zhong C, Wang S, Lai H, Guo Y, Lv D, Lu J, Mao X. Gut dysbiosis promotes prostate cancer progression and docetaxel resistance via activating NF-κB-IL6-STAT3 axis. Microbiome. 2022;10:94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Gil da Costa RM, Levesque C, Bianchi-Frias D, Chatterjee P, Lam HM, Santos C, Coleman IM, Ferreirinha P, Vilanova M et al. Pinto da Cunha N, : Pharmacological NF-κB inhibition decreases cisplatin chemoresistance in muscle-invasive bladder cancer and reduces cisplatin-induced toxicities. Mol Oncol 2023, 17:2709–2727.

  202. Jin X, Ding D, Yan Y, Li H, Wang B, Ma L, Ye Z, Ma T, Wu Q, Rodrigues DN, et al. Phosphorylated RB promotes Cancer immunity by inhibiting NF-κB activation and PD-L1 expression. Mol Cell. 2019;73:22–e3526.

    Article  PubMed  CAS  Google Scholar 

  203. Li WF, Herkilini A, Tang Y, Huang P, Song GB, Miyagishi M, Kasim V, Wu SR. The transcription factor PBX3 promotes tumor cell growth through transcriptional suppression of the tumor suppressor p53. Acta Pharmacol Sin. 2021;42:1888–99.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Wang R, Liu X. Epigenetic regulation of prostate cancer. Genes Dis. 2020;7:606–13.

    Article  PubMed  CAS  Google Scholar 

  205. Huang Y, Lu S, Chen Y, Qing Y, Wu R, Ma T, Zhang Z, Wang Y, Li K. Verification of cell cycle-associated cyclin-dependent kinases facilitated prostate cancer progression by integrated bioinformatic analysis and experimental validation. Heliyon. 2022;8:e10081.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  206. Bar-Ziv R, Brodsky S, Chapal M, Barkai N. Transcription factor binding to replicated DNA. Cell Rep. 2020;30:3989–e39953984.

    Article  PubMed  CAS  Google Scholar 

  207. Atsaves V, Leventaki V, Rassidakis GZ, Claret FX. AP-1 transcription factors as regulators of Immune responses in cancer. Cancers (Basel). 2019;11:1037.

  208. Luo CT, Li MO. Foxo transcription factors in T cell biology and tumor immunity. Semin Cancer Biol. 2018;50:13–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  209. Wang Y, Huang Z, Sun M, Huang W, Xia L. ETS transcription factors: multifaceted players from cancer progression to tumor immunity. Biochim Biophys Acta Rev Cancer. 2023;1878:188872.

    Article  PubMed  CAS  Google Scholar 

  210. Su Y, Zhang Y, Zhao J, Zhou W, Wang W, Han B, Wang X. FOXA1 promotes prostate cancer angiogenesis by inducing multiple pro-angiogenic factors expression. J Cancer Res Clin Oncol. 2021;147:3225–43.

    Article  PubMed  CAS  Google Scholar 

  211. Miettinen M, Wang ZF, Paetau A, Tan SH, Dobi A, Srivastava S, Sesterhenn I. ERG transcription factor as an immunohistochemical marker for vascular endothelial tumors and prostatic carcinoma. Am J Surg Pathol. 2011;35:432–41.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Sanmukh SG, Dos Santos NJ, Nascimento Barquilha C, De Carvalho M, Pintor Dos Reis P, Delella FK, Carvalho HF, Latek D, Fehér T, Felisbino SL. Bacterial RNA virus MS2 exposure increases the expression of cancer progression genes in the LNCaP prostate cancer cell line. Oncol Lett. 2023;25:86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–7.

    Article  PubMed  CAS  Google Scholar 

  214. Peiffer LB, White JR, Jones CB, Slottke RE, Ernst SE, Moran AE, Graff JN, Sfanos KS. Composition of gastrointestinal microbiota in association with treatment response in individuals with metastatic castrate resistant prostate cancer progressing on enzalutamide and initiating treatment with anti-PD-1 (pembrolizumab). Neoplasia. 2022;32:100822.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  215. Mirzaei R, Afaghi A, Babakhani S, Sohrabi MR, Hosseini-Fard SR, Babolhavaeji K, Khani Ali Akbari S, Yousefimashouf R, Karampoor S. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed Pharmacother. 2021;139:111619.

    Article  PubMed  CAS  Google Scholar 

  216. Li JKM, Wang LL, Wong CYP, Chiu PKF, Teoh JYC, Kwok HSW, Leung SCH, Wong SH, Tsui SKW, Ng CF. A cross-sectional study on gut microbiota in prostate cancer patients with prostatectomy or androgen deprivation therapy. Prostate Cancer Prostatic Dis. 2021;24:1063–72.

    Article  PubMed  CAS  Google Scholar 

  217. Lachance G, Robitaille K, Laaraj J, Gevariya N, Varin TV, Feldiorean A, Gaignier F, Julien IB, Xu HW, Hallal T, et al. The gut microbiome-prostate cancer crosstalk is modulated by dietary polyunsaturated long-chain fatty acids. Nat Commun. 2024;15:3431.

  218. Terrisse S, Zitvogel L, Kroemer G. Effects of the intestinal microbiota on prostate cancer treatment by androgen deprivation therapy. Microb Cell. 2022;9:202–6.

    Article  PubMed  Google Scholar 

  219. Terrisse S, Goubet AG, Ueda K, Thomas AM, Quiniou V, Thelemaque C, Dunsmore G, Clave E, Gamat-Huber M, Yonekura S et al. Immune system and intestinal microbiota determine efficacy of androgen deprivation therapy against prostate cancer. J Immunother Cancer. 2022;10:e004191.

  220. Mavundza EJ, Mmotsa TM, Ndwandwe D. Human papillomavirus (HPV) trials: a cross-sectional analysis of clinical trials registries. Hum Vaccin Immunother. 2024;20:2393481.

    Article  PubMed  PubMed Central  Google Scholar 

  221. Russo GI, Calogero AE, Condorelli RA, Scalia G, Morgia G, La Vignera S. Human papillomavirus and risk of prostate cancer: a systematic review and meta-analysis. Aging Male. 2020;23:132–8.

    Article  PubMed  Google Scholar 

  222. Brimo F, Montironi R, Egevad L, Erbersdobler A, Lin DW, Nelson JB, Rubin MA, van der Kwast T, Amin M, Epstein JI. Contemporary grading for prostate cancer: implications for patient care. Eur Urol. 2013;63:892–901.

    Article  PubMed  Google Scholar 

  223. Nahand JS, Khanaliha K, Mirzaei H, Moghoofei M, Baghi HB, Esghaei M, Khatami AR, Fatemipour M, Bokharaei-Salim F. Possible role of HPV/EBV coinfection in anoikis resistance and development in prostate cancer. BMC Cancer. 2021;21:926.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  224. Sufiawati I, Harmiyati R, Nur’aeny N, Indrati AR, Lesmana R, Wisaksana R, Amalia R. Detection of human herpesviruses in sera and saliva of asymptomatic HIV-infected individuals using multiplex RT-PCR DNA microarray. Pathogens. 2023;12:993.

  225. Wang L, Ning J, Wakimoto H, Wu S, Wu CL, Humphrey MR, Rabkin SD, Martuza RL. Oncolytic herpes simplex virus and PI3K inhibitor BKM120 synergize to promote killing of prostate Cancer stem-like cells. Mol Ther Oncolytics. 2019;13:58–66.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Sutcliffe S, Till C, Jenkins FJ, Gaydos CA, Goodman PJ, Hoque AM, Hsing AW, Thompson IM, Nelson WG, De Marzo AM, Platz EA. Prospective study of human herpesvirus type 8 serostatus and prostate cancer risk in the placebo arm of the prostate Cancer Prevention Trial. Cancer Causes Control. 2015;26:35–44.

    Article  PubMed  Google Scholar 

  227. Bahmad HF, Jalloul M, Azar J, Moubarak MM, Samad TA, Mukherji D, Al-Sayegh M, Abou-Kheir W. Tumor microenvironment in prostate Cancer: toward identification of novel molecular biomarkers for diagnosis, prognosis, and Therapy Development. Front Genet. 2021;12:652747.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  228. Bussemakers MJ, Van Bokhoven A, Tomita K, Jansen CF, Schalken JA. Complex cadherin expression in human prostate cancer cells. Int J Cancer. 2000;85:446–50.

    Article  PubMed  CAS  Google Scholar 

  229. Gong Y, Chippada-Venkata UD, Oh WK. Roles of matrix metalloproteinases and their natural inhibitors in prostate cancer progression. Cancers (Basel). 2014;6:1298–327.

    Article  PubMed  Google Scholar 

  230. Jiao M, Qi M, Zhang F, Hu J, Feng T, Zhao M, Li X, Liu H, Teng W, Zhang J, et al. CUL4B regulates cancer stem-like traits of prostate cancer cells by targeting BMI1 via miR200b/c. Prostate. 2019;79:1294–303.

    Article  PubMed  CAS  Google Scholar 

  231. Hu J, Sun F, Chen W, Zhang J, Zhang T, Qi M, Feng T, Liu H, Li X, Xing Y, et al. BTF3 sustains cancer stem-like phenotype of prostate cancer via stabilization of BMI1. J Exp Clin Cancer Res. 2019;38:227.

    Article  PubMed  PubMed Central  Google Scholar 

  232. Simeckova S, Kahounova Z, Fedr R, Remsik J, Slabakova E, Suchankova T, Prochazkova J, Bouchal J, Kharaishvili G, Kral M, et al. High Skp2 expression is associated with a mesenchymal phenotype and increased tumorigenic potential of prostate cancer cells. Sci Rep. 2019;9:5695.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Yoo YA, Vatapalli R, Lysy B, Mok H, Desouki MM, Abdulkadir SA. The role of castration-resistant Bmi1 + Sox2 + cells in driving recurrence in prostate Cancer. J Natl Cancer Inst. 2019;111:311–21.

    Article  PubMed  Google Scholar 

  234. Federer-Gsponer JR, Muller DC, Zellweger T, Eggimann M, Marston K, Ruiz C, Seifert HH, Rentsch CA, Bubendorf L, Le Magnen C. Patterns of stemness-associated markers in the development of castration-resistant prostate cancer. Prostate. 2020;80:1108–17.

    Article  PubMed  CAS  Google Scholar 

  235. Lee CH, Jeon YT, Kim SH, Song YS. NF-kappaB as a potential molecular target for cancer therapy. BioFactors. 2007;29:19–35.

    Article  PubMed  CAS  Google Scholar 

  236. Culig Z, Puhr M. Interleukin-6 and prostate cancer: current developments and unsolved questions. Mol Cell Endocrinol. 2018;462:25–30.

    Article  PubMed  CAS  Google Scholar 

  237. Lopez-Bujanda ZA, Haffner MC, Chaimowitz MG, Chowdhury N, Venturini NJ, Patel RA, Obradovic A, Hansen CS, Jackow J, Maynard JP, et al. Castration-mediated IL-8 promotes myeloid infiltration and prostate cancer progression. Nat Cancer. 2021;2:803–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  238. Kloss CC, Lee J, Zhang A, Chen F, Melenhorst JJ, Lacey SF, Maus MV, Fraietta JA, Zhao Y, June CH. Dominant-negative TGF-beta receptor enhances PSMA-Targeted human CAR T cell proliferation and augments prostate Cancer eradication. Mol Ther. 2018;26:1855–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  239. Akl MR, Nagpal P, Ayoub NM, Tai B, Prabhu SA, Capac CM, Gliksman M, Goy A, Suh KS. Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget. 2016;7:44735–62.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Chen J, De S, Brainard J, Byzova TV. Metastatic properties of prostate cancer cells are controlled by VEGF. Cell Commun Adhes. 2004;11:1–11.

    Article  PubMed  Google Scholar 

  241. Stallone G, Cormio L, Netti GS, Infante B, Selvaggio O, Fino GD, Ranieri E, Bruno F, Prattichizzo C, Sanguedolce F, et al. Pentraxin 3: a novel biomarker for predicting progression from prostatic inflammation to prostate cancer. Cancer Res. 2014;74:4230–8.

    Article  PubMed  CAS  Google Scholar 

  242. Wang W, Bergh A, Damber JE. Cyclooxygenase-2 expression correlates with local chronic inflammation and tumor neovascularization in human prostate cancer. Clin Cancer Res. 2005;11:3250–6.

    Article  PubMed  CAS  Google Scholar 

  243. Satoh N, Yamada Y, Kinugasa Y, Takakura N. Angiopoietin-1 alters tumor growth by stabilizing blood vessels or by promoting angiogenesis. Cancer Sci. 2008;99:2373–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  244. Chang YS, Chen WY, Yin JJ, Sheppard-Tillman H, Huang J, Liu YN. EGF receptor promotes prostate Cancer bone metastasis by downregulating miR-1 and activating TWIST1. Cancer Res. 2015;75:3077–86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  245. Jung Y, Wang J, Schneider A, Sun YX, Koh-Paige AJ, Osman NI, McCauley LK, Taichman RS. Regulation of SDF-1 (CXCL12) production by osteoblasts; a possible mechanism for stem cell homing. Bone. 2006;38:497–508.

    Article  PubMed  CAS  Google Scholar 

  246. Hou Y, Li H, Huo W. THBS4 silencing regulates the cancer stem cell-like properties in prostate cancer via blocking the PI3K/Akt pathway. Prostate. 2020;80:753–63.

    Article  PubMed  CAS  Google Scholar 

  247. Sun D, Wang J, Han Y, Dong X, Ge J, Zheng R, Shi X, Wang B, Li Z, Ren P, et al. TISCH: a comprehensive web resource enabling interactive single-cell transcriptome visualization of tumor microenvironment. Nucleic Acids Res. 2021;49:D1420–30.

    Article  PubMed  CAS  Google Scholar 

  248. Al Shareef Z, Hachim MY, Bouzid A, Talaat IM, Al-Rawi N, Hamoudi R, Hachim IY. The prognostic value of Dickkopf-3 (Dkk3), TGFB1 and ECM-1 in prostate cancer. Front Mol Biosci. 2024;11:1351888.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  249. Crescitelli R, Lasser C, Lotvall J. Isolation and characterization of extracellular vesicle subpopulations from tissues. Nat Protoc. 2021;16:1548–80.

    Article  PubMed  CAS  Google Scholar 

  250. Chiarugi P, Paoli P, Cirri P. Tumor microenvironment and metabolism in prostate cancer. Semin Oncol. 2014;41:267–80.

    Article  PubMed  CAS  Google Scholar 

  251. Altuna-Coy A, Ruiz-Plazas X, Sanchez-Martin S, Ascaso-Til H, Prados-Saavedra M, Alves-Santiago M, Bernal-Escote X, Segarra-Tomas J. M RC: The lipidomic profile of the tumoral periprostatic adipose tissue reveals alterations in tumor cell’s metabolic crosstalk. BMC Med 2022, 20:255.

  252. Ding Y, Lee M, Gao Y, Bu P, Coarfa C, Miles B, Sreekumar A, Creighton CJ, Ayala G. Neuropeptide Y nerve paracrine regulation of prostate cancer oncogenesis and therapy resistance. Prostate. 2021;81:58–71.

    Article  PubMed  CAS  Google Scholar 

  253. Lodyga M, Hinz B. TGF-beta1 - A truly transforming growth factor in fibrosis and immunity. Semin Cell Dev Biol. 2020;101:123–39.

    Article  PubMed  CAS  Google Scholar 

  254. Barnestein R, Galland L, Kalfeist L, Ghiringhelli F, Ladoire S, Limagne E. Immunosuppressive tumor microenvironment modulation by chemotherapies and targeted therapies to enhance immunotherapy effectiveness. Oncoimmunology. 2022;11:2120676.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Jarvis EM, Collings S, Authier-Hall A, Dasyam N, Luey B, Nacey J, Painter GF, Delahunt B, Hermans IF, Weinkove R. Mucosal-Associated Invariant T (MAIT) cell dysfunction and PD-1 expression in prostate Cancer: implications for Immunotherapy. Front Immunol. 2021;12:748741.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  256. Isaacsson Velho P, Antonarakis ES. PD-1/PD-L1 pathway inhibitors in advanced prostate cancer. Expert Rev Clin Pharmacol. 2018;11:475–86.

    Article  PubMed  CAS  Google Scholar 

  257. Santoni M, Massari F, Cheng L, Cimadamore A, Scarpelli M, Montironi R, Lopez-Beltran A. PD-L1 inhibitors for the treatment of prostate Cancer. Curr Drug Targets. 2020;21:1558–65.

    Article  PubMed  CAS  Google Scholar 

  258. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501:328–37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  259. Niu M, Yi M, Wu Y, Lyu L, He Q, Yang R, Zeng L, Shi J, Zhang J, Zhou P, et al. Synergistic efficacy of simultaneous anti-TGF-beta/VEGF bispecific antibody and PD-1 blockade in cancer therapy. J Hematol Oncol. 2023;16:94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Batlle E, Massague J. Transforming growth factor-beta signaling in immunity and Cancer. Immunity. 2019;50:924–40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  261. Ali S, Rehman MU, Yatoo AM, Arafah A, Khan A, Rashid S, Majid S, Ali A, Ali MN. TGF-beta signaling pathway: therapeutic targeting and potential for anti-cancer immunity. Eur J Pharmacol. 2023;947:175678.

    Article  PubMed  CAS  Google Scholar 

  262. Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in Human Disease. Cell. 2017;170:605–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  263. Da Silva CG, Camps MGM, Li T, Chan AB, Ossendorp F, Cruz LJ. Co-delivery of immunomodulators in biodegradable nanoparticles improves therapeutic efficacy of cancer vaccines. Biomaterials. 2019;220:119417.

    Article  PubMed  Google Scholar 

  264. Eygeris Y, Gupta M, Kim J, Sahay G. Chemistry of lipid nanoparticles for RNA delivery. Acc Chem Res. 2022;55:2–12.

    Article  PubMed  CAS  Google Scholar 

  265. Borges RC, Tourinho-Barbosa RR, de la Rosette J. Tumour microenvironment and focal therapy for prostate cancer. Curr Opin Urol. 2022;32:248–53.

    Article  PubMed  Google Scholar 

  266. Dai J, Lu Y, Roca H, Keller JM, Zhang J, McCauley LK, Keller ET. Immune mediators in the tumor microenvironment of prostate cancer. Chin J Cancer. 2017;36:29.

    Article  PubMed  PubMed Central  Google Scholar 

  267. Nagireddy S, Qureshi R, Best J, Frech FS, Shah K, Soni Y, Kuchakulla M, Narasimman M, Arora H. Current treatment modalities targeting Tumor Microenvironment in Castration-resistant prostate Cancer. Adv Exp Med Biol. 2021;1329:295–323.

    Article  PubMed  CAS  Google Scholar 

  268. Muntimadugu E, Kommineni N, Khan W. Exploring the potential of Nanotherapeutics in Targeting Tumor Microenvironment for Cancer Therapy. Pharmacol Res. 2017;126:109–22.

    Article  PubMed  CAS  Google Scholar 

  269. Cheng B, Tang C, Xie J, Zhou Q, Luo T, Wang Q, Huang H. Cuproptosis illustrates tumor micro-environment features and predicts prostate cancer therapeutic sensitivity and prognosis. Life Sci. 2023;325:121659.

    Article  PubMed  CAS  Google Scholar 

  270. Cheng YT, Yang CC, Shyur LF. Phytomedicine-modulating oxidative stress and the tumor microenvironment for cancer therapy. Pharmacol Res. 2016;114:128–43.

    Article  PubMed  Google Scholar 

  271. Messex JK, Liou GY. Impact of immune cells in the tumor microenvironment of prostate cancer metastasis. Life (Basel). 2023;13:333.

  272. Petrylak DP, Ratta R, Gafanov R, Facchini G, Piulats JM, Kramer G, Flaig TW, Chandana SR, Li B, Burgents J, Fizazi K. KEYNOTE-921: phase III study of pembrolizumab plus docetaxel for metastatic castration-resistant prostate cancer. Future Oncol. 2021;17:3291–9.

    Article  PubMed  CAS  Google Scholar 

  273. Graff JN, Alumkal JJ, Drake CG, Thomas GV, Redmond WL, Farhad M, Cetnar JP, Ey FS, Bergan RC, Slottke R, Beer TM. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget. 2016;7:52810–7.

    Article  PubMed  PubMed Central  Google Scholar 

  274. Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, Vaishampayan U, Berger R, Sezer A, Alanko T, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate Cancer: Multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol. 2020;38:395–405.

    Article  PubMed  CAS  Google Scholar 

  275. Alaia C, Boccellino M, Zappavigna S, Amler E, Quagliuolo L, Rossetti S, Facchini G, Caraglia M. Ipilimumab for the treatment of metastatic prostate cancer. Expert Opin Biol Ther. 2018;18:205–13.

    Article  PubMed  CAS  Google Scholar 

  276. Hussain M, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, Chi KN, Sartor O, Agarwal N, Olmos D, et al. Survival with Olaparib in Metastatic Castration-resistant prostate Cancer. N Engl J Med. 2020;383:2345–57.

    Article  PubMed  CAS  Google Scholar 

  277. de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, Chi KN, Sartor O, Agarwal N, Olmos D, et al. Olaparib for metastatic castration-resistant prostate Cancer. N Engl J Med. 2020;382:2091–102.

    Article  PubMed  Google Scholar 

  278. Barata PC, Cooney M, Mendiratta P, Gupta R, Dreicer R, Garcia JA. Phase I/II study evaluating the safety and clinical efficacy of temsirolimus and bevacizumab in patients with chemotherapy refractory metastatic castration-resistant prostate cancer. Invest New Drugs. 2019;37:331–7.

    Article  PubMed  CAS  Google Scholar 

  279. Zhao F, Tian W, Zeng M, Xia J, Hu H, Hao X, Han L, Liu H, He Y, Zhu X, et al. Apatinib alone or combined with radiotherapy in metastatic prostate cancer: results from a pilot, multicenter study. Oncotarget. 2017;8:110774–84.

    Article  PubMed  PubMed Central  Google Scholar 

  280. Ward JE, Karrison T, Chatta G, Hussain M, Shevrin D, Szmulewitz RZ, O’Donnell PH, Stadler WM, Posadas EM. A randomized, phase II study of pazopanib in castrate-sensitive prostate cancer: a University of Chicago Phase II Consortium/Department of Defense Prostate Cancer Clinical Trials Consortium study. Prostate Cancer Prostatic Dis. 2012;15:87–92.

    Article  PubMed  CAS  Google Scholar 

  281. Ryan CJ, Stadler WM, Roth B, Hutcheon D, Conry S, Puchalski T, Morris C, Small EJ. Phase I dose escalation and pharmacokinetic study of AZD2171, an inhibitor of the vascular endothelial growth factor receptor tyrosine kinase, in patients with hormone refractory prostate cancer (HRPC). Invest New Drugs. 2007;25:445–51.

    Article  PubMed  CAS  Google Scholar 

  282. Saylor PJ, Mahmood U, Kunawudhi A, Smith MR, Palmer EL, Michaelson MD. Multitargeted tyrosine kinase inhibition produces discordant changes between 99mTc-MDP bone scans and other disease biomarkers: analysis of a phase II study of sunitinib for metastatic castration-resistant prostate cancer. J Nucl Med. 2012;53:1670–5.

    Article  PubMed  CAS  Google Scholar 

  283. Nakamura M, Ikeda M, Komukai M, Suyemitsu T, Okinaga S, Arai K. Presence of a 16 kd protein in human seminal plasma counteracts the effects of the anti-fertility agent, gossypol. Hum Reprod. 1991;6:714–21.

    Article  PubMed  CAS  Google Scholar 

  284. Liu G, Kelly WK, Wilding G, Leopold L, Brill K, Somer B. An open-label, multicenter, phase I/II study of single-agent AT-101 in men with castrate-resistant prostate cancer. Clin Cancer Res. 2009;15:3172–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  285. Huang G, Zhang H, Shi H, Zhang W, Wang T, Wang Z, Chen Q, Lian B, Li J, Yang G. Clinicopathological and immunological profiles of prostate adenocarcinoma and neuroendocrine prostate cancer. World J Surg Oncol. 2022;20:407.

    Article  PubMed  PubMed Central  Google Scholar 

  286. Droz JP, Medioni J, Chevreau C, De Mont-Serrat H, Merger M, Stopfer P, Kaiser R, Oudard S. Randomized phase II study of nintedanib in metastatic castration-resistant prostate cancer postdocetaxel. Anticancer Drugs. 2014;25:1081–8.

    Article  PubMed  CAS  Google Scholar 

  287. Izumi K, Mizokami A, Shima T, Narimoto K, Sugimoto K, Kobori Y, Maeda Y, Konaka H, Koh E, Namiki M. Preliminary results of tranilast treatment for patients with advanced castration-resistant prostate cancer. Anticancer Res. 2010;30:3077–81.

    PubMed  Google Scholar 

  288. Mori JO, Elhussin I, Brennen WN, Graham MK, Lotan TL, Yates CC, De Marzo AM, Denmeade SR, Yegnasubramanian S, Nelson WG, et al. Prognostic and therapeutic potential of senescent stromal fibroblasts in prostate cancer. Nat Rev Urol. 2024;21:258–73.

    Article  PubMed  Google Scholar 

  289. Stein MN, Hussain M, Stadler WM, Liu G, Tereshchenko IV, Goodin S, Jeyamohan C, Kaufman HL, Mehnert J, DiPaola RS. A phase II study of AT-101 to overcome Bcl-2–Mediated resistance to Androgen Deprivation Therapy in patients with newly diagnosed castration-sensitive metastatic prostate Cancer. Clin Genitourin Cancer. 2016;14:22–7.

    Article  PubMed  Google Scholar 

  290. Zha C, Peng Z, Huang K, Tang K, Wang Q, Zhu L, Che B, Li W, Xu S, Huang T, et al. Potential role of gut microbiota in prostate cancer: immunity, metabolites, pathways of action? Front Oncol. 2023;13:1196217.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  291. Pernigoni N, Zagato E, Calcinotto A, Troiani M, Mestre RP, Cali B, Attanasio G, Troisi J, Minini M, Mosole S, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374:216–24.

    Article  PubMed  CAS  Google Scholar 

  292. El Tekle G, Garrett WS. Bacteria in cancer initiation, promotion and progression. Nat Rev Cancer. 2023;23:600–18.

    Article  PubMed  Google Scholar 

  293. Campbell CT, Gulley JL, Oyelaran O, Hodge JW, Schlom J, Gildersleeve JC. Humoral response to a viral glycan correlates with survival on PROSTVAC-VF. Proc Natl Acad Sci U S A. 2014;111:E1749–1758.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  294. Madan RA, Heery CR, Gulley JL. Poxviral-based vaccine elicits immunologic responses in prostate cancer patients. Oncoimmunology. 2014;3:e28611.

    Article  PubMed  PubMed Central  Google Scholar 

  295. McNeel DG, Eickhoff JC, Johnson LE, Roth AR, Perk TG, Fong L, Antonarakis ES, Wargowski E, Jeraj R, Liu G. Phase II trial of a DNA vaccine Encoding Prostatic Acid Phosphatase (pTVG-HP [MVI-816]) in patients with Progressive, Nonmetastatic, castration-sensitive prostate Cancer. J Clin Oncol. 2019;37:3507–17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  296. De Santis M. [Immunotherapy for the treatment of prostate cancer-a comeback?]. Urologe A. 2018;57:1342–5.

    PubMed  Google Scholar 

  297. Madan RA, Gulley JL, Kantoff PW. Demystifying immunotherapy in prostate cancer: understanding current and future treatment strategies. Cancer J. 2013;19:50–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  298. Adhyatma KP, Prapiska FF, Siregar GP, Warli SM. Systemic inflammatory response in Predicting prostate Cancer: the diagnostic value of Neutrophil-To-Lymphocyte ratio. Open Access Maced J Med Sci. 2019;7:1628–30.

    Article  PubMed  PubMed Central  Google Scholar 

  299. Lautert-Dutra W, C MM, Chaves LP, Crozier C, F PS, Bayani RBDR, Bonatto J, Squire SL. Loss of heterozygosity impacts MHC expression on the immune microenvironment in CDK12-mutated prostate cancer. Mol Cytogenet. 2024;17:11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  300. Fang B, Wei Y, Zeng H, Li Y, Chen S, Zhang T, Pan J, Wang B, Wu J, Jin S, et al. Prevalence of mismatch repair genes mutations and clinical activity of PD-1 therapy in Chinese prostate cancer patients. Cancer Immunol Immunother. 2023;72:1541–51.

    Article  PubMed  CAS  Google Scholar 

  301. Zhou R, Lu D, Mi J, Wang C, Lu W, Wang Z, Li X, Wei C, Zhang H, Ji J, et al. Disulfidptosis-related genes serve as potential prognostic biomarkers and indicate tumor microenvironment characteristics and immunotherapy response in prostate cancer. Sci Rep. 2024;14:14107.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  302. Hougen HY, Graf RP, Li G, Quintanilha JCF, Lin DI, Ross JS, Punnen S, Mahal BA. Clinical and Genomic Factors Associated with Greater Tumor Mutational Burden in prostate Cancer. Eur Urol Open Sci. 2023;55:45–9.

    Article  PubMed  PubMed Central  Google Scholar 

  303. Graf RP, Fisher V, Weberpals J, Gjoerup O, Tierno MB, Huang RSP, Sayegh N, Lin DI, Raskina K, Schrock AB, et al. Comparative effectiveness of Immune checkpoint inhibitors vs chemotherapy by Tumor Mutational Burden in Metastatic Castration-resistant prostate Cancer. JAMA Netw Open. 2022;5:e225394.

    Article  PubMed  PubMed Central  Google Scholar 

  304. Iannantuono GM, Torino F, Rosenfeld R, Guerriero S, Carlucci M, Sganga S, Capotondi B, Riondino S, Roselli M. The role of histology-agnostic drugs in the treatment of metastatic castration-resistant prostate cancer. Int J Mol Sci. 2022;23:8535.

  305. Lenis AT, Ravichandran V, Brown S, Alam SM, Katims A, Truong H, Reisz PA, Vasselman S, Nweji B, Autio KA, et al. Microsatellite instability, Tumor Mutational Burden, and response to Immune Checkpoint Blockade in patients with prostate Cancer. Clin Cancer Res. 2024;30:3894–903.

    Article  PubMed  Google Scholar 

  306. Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, Rathkopf D, Morris MJ, Danila DC, Slovin SF, et al. Analysis of the prevalence of microsatellite instability in prostate Cancer and response to Immune Checkpoint Blockade. JAMA Oncol. 2019;5:471–8.

    Article  PubMed  Google Scholar 

  307. Wu YM, Cieslik M, Lonigro RJ, Vats P, Reimers MA, Cao X, Ning Y, Wang L, Kunju LP, de Sarkar N, et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate Cancer. Cell. 2018;173:1770–e17821714.

    Article  PubMed  CAS  Google Scholar 

  308. Antonarakis ES, Isaacsson Velho P, Fu W, Wang H, Agarwal N, Sacristan Santos V, Maughan BL, Pili R, Adra N, Sternberg CN, et al. CDK12-Altered prostate Cancer: clinical features and therapeutic outcomes to standard systemic therapies, poly (ADP-Ribose) polymerase inhibitors, and PD-1 inhibitors. JCO Precis Oncol. 2020;4:370–81.

    Article  PubMed  Google Scholar 

  309. Guo J, Fang J, Huang X, Liu Y, Yuan Y, Zhang X, Zou C, Xiao K, Wang J. Prognostic role of neutrophil to lymphocyte ratio and platelet to lymphocyte ratio in prostate cancer: a meta-analysis of results from multivariate analysis. Int J Surg. 2018;60:216–23.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The figures were prepared using www.BioRender.com.

Funding

This work was financially supported by the National Natural Science Foundation of China (No. 82074270).

Author information

Authors and Affiliations

Authors

Contributions

Lin Chen: investigation, writing original-draft, drawing. Yu-Xin Xu: drawing, check. Yuan-Shuo Wang: writing original-draft, table. Ying-Ying Ren: writing original-draft, check. Xue-Man Dong: writing original-draft. Pu Wu: writing original-draft. Tian Xie, Qi Zhang, Jian-Liang Zhou: conceptualization, project administration, writing-review & editing.

Corresponding authors

Correspondence to Tian Xie, Qi Zhang or Jian-Liang Zhou.

Ethics declarations

Ethical approval

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, L., Xu, YX., Wang, YS. et al. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 23, 229 (2024). https://doi.org/10.1186/s12943-024-02137-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12943-024-02137-1

Keywords