Download PDF
Review  |  Open Access  |  23 Jul 2024

The reactive stroma response regulates the immune landscape in prostate cancer

Views: 78 |  Downloads: 26 |  Cited:  0
J Transl Genet Genom 2024;8:249-77.
10.20517/jtgg.2024.15 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Prostate cancer remains the most commonly diagnosed and the second leading cause of cancer-related deaths in men in the United States. The neoplastic transformation of prostate epithelia, concomitant with modulations in the stromal compartment, known as reactive stromal response, is critical for the growth, development, and progression of prostate cancer. Reactive stroma typifies an emergent response to disrupted tissue homeostasis commonly observed in wound repair and pathological conditions such as cancer. Despite the significance of reactive stroma in prostate cancer pathobiology, our understanding of the ontogeny, phenotypic and functional heterogeneity, and reactive stromal regulation of the immune microenvironment in prostate cancer remains limited. Traditionally characterized to have an immunologically "cold" tumor microenvironment, prostate cancer presents significant challenges for advancing immunotherapy compared to other solid tumors. This review explores the detrimental role of reactive stroma in prostate cancer, particularly its immunomodulatory function. Understanding the molecular characteristics and dynamic transcriptional program of the reactive stromal populations in tandem with tumor progression could offer insights into enhancing immunotherapy efficacy against prostate cancer.

Keywords

Prostate cancer, emergent/repair biology, reactive stroma, tumor microenvironment, immune landscape, immunotherapy

INTRODUCTION

The prostate, an exocrine gland located beneath the urinary bladder and surrounding the urethra, is essential for male reproductive function. During embryonic development, epithelial buds from the urogenital sinus interact with mesenchyme, driving prostate differentiation and secretory duct formation[1-3]. Cancer of the prostate gland, the most diagnosed cancer in men and the second-leading cause of male cancer deaths in the United States, typically progresses slowly[4-6]. Prostate cancer incidence and mortality rates are closely linked to aging. Additionally, in the United States, African American men have a higher incidence rate of developing aggressive prostate cancer compared to their Caucasian counterparts[4,7]. While localized disease responds well to surgery or radiation, 20% to 50% of patients experience biochemical recurrence within a decade, leading to advanced or metastatic cancer[7]. The prostate stroma plays a critical role in promoting the progression of advanced prostate cancer[8].

In adult differentiated biology, the physical and biochemical interactions between the epithelial cells together with cellular and non-cellular components of the stroma regulate normal prostate function and homeostasis[9]. The prostate stroma is composed of smooth muscle cells, tissue-resident mesenchymal cells, extracellular matrix (ECM) proteins, nerves, blood vessels, and a spectrum of immune cells [Figure 1][9-11]. During development, androgen secreted by the testis is the chief regulator of prostate gland growth, and the hormonal action is mediated through the androgen receptor (AR)[12]. However, tissue recombination experiments have revealed that the androgenic effects on prostate gland growth and development are not solely dependent on epithelial AR but require paracrine signaling induced by the AR-positive prostate mesenchyme[13,14]. Disruptions in the functional coupling between the stromal and epithelial interactions in the adult prostate are associated with glandular dysfunction typified in aging and disease, although specific mechanisms remain undefined. Considerable evidence suggests these biological disruptions are associated with a transition into a repair-centric, emergency, or emergent tissue systems biology state to affect rapid repair until the biological priority resets to functional, differentiated biology[15]. However, an unresolved/chronic repair state of the stroma, defined as reactive stroma, has been correlated with promoting the evolution of pathological processes including benign prostate hyperplasia (BPH) and prostate cancer[15-18] [Figure 1].

The reactive stroma response regulates the immune landscape in prostate cancer

Figure 1. Reactive/emergent stromal response in prostate. (A) The prostate gland at homeostasis maintains a well-organized tissue architecture with specific cellular components functioning in a balanced state. (B) Aging and conditions like benign prostatic hyperplasia (BPH) and cancer disrupt this homeostasis, triggering an emergency/emergent (repair) processes in the prostate tissue to restore homeostasis.

Currently, there is very limited information available on both tissue-resident immune cells in healthy adult prostate and the modulations that occur in the immune landscape in a disease state like cancer. Often co-evolving with cancer, the reactive stroma is an environment enriched with growth factors and characterized by increased angiogenesis, an increased inflammatory response, and an extensively remodeled ECM resulting in a desmoplastic reaction[11,19]. The reactive stroma of solid tumors, including prostate cancer, has been shown to be immunosuppressive and associated with induced resistance to tumor-targeted immunotherapies; however, the mechanisms remain complex[20,21]. Recent studies have shown that either reprogramming subsets of stromal cells or immunotherapies targeting stromal antigens can disrupt the pro-tumorigenic microenvironment niche and enhance endogenous or vaccine-induced antitumor immunity[20-24]. In this review, we will discuss the current knowledge about stromal evolution in prostate cancer tumorigenesis and its known regulation of the tissue-immune landscape. Understanding this will help in developing effective therapeutic strategies that can be leveraged to co-target peri-tumoral reactive stroma to reprogram the immune suppressive tumor microenvironment (TME) and render it permissive to antitumor immunotherapies.

PROSTATE GLAND AT HOMEOSTASIS

An integrative network involving various cellular and acellular components regulates the structure, function, and homeostasis of the prostate gland. The acinar epithelial cells constitute the functional parenchyma of the prostate gland. The stromal cells synthesize ECM components and provide mechanical support to the secretory epithelium. In addition to these functions, immune cells actively participate in the surveillance of organ integrity, while the vascular system provides oxygen and nutrient support to the organ. Moreover, the contractile activity of the smooth muscle of the stroma is pivotal for the proper functioning of the prostate gland. This activity is regulated by neuronal inputs, with the sympathetic nervous system acting via the hypogastric nerve, and parasympathetic nervous system via the pelvic nerve[1,2,25].

The bulk stroma of the prostate consists of fibroblasts, mesenchymal stem cells, and smooth muscle cells. The homeostasis and normal functioning of the prostate gland are dependent on intercellular communications between the epithelial and stromal compartment, mediated through the paracrine and apocrine secretions from both cell types[25-30]. Various secretory effectors like wingless-related integration sites (WNTs), transforming growth factors (TGF) α and β, fibroblast growth factors (FGF), insulin growth factors (IGF), epidermal growth factor (EGF), platelet-derived growth factors (PDGF), vascular endothelial growth factor (VEGF), prostaglandins, endothelin, sonic hedgehog, and nitrous oxide orchestrate cellular proliferation, differentiation, and the regulation of cell death in both epithelial and stromal cells of the prostate gland. These processes are mediated via the respective cognate receptors[27,30,31]. Furthermore, a well-defined laminin-positive basement membrane demarcates the epithelial acini from the fibromuscular stroma of the prostate gland[32]. Along with key junctional complexes within the epithelium, the structural integrity of this basement membrane is crucial for maintaining the functionality of the acini and preventing the dissemination of pathogenic microbes into the stromal tissue. A breach in basement membrane integrity also serves as a critical precursor to invasive progression and systemic pathogenesis like prostate cancer metastases[33,34].

Steroid regulation of the prostate

Steroids, particularly testosterone, play an important role in maintaining the structural and functional integrity of the prostate gland. At the subcellular level, androstenedione is also essential for maintaining the function of prostatic epithelium. In addition to androgens, estrogen contributes to the regulation of prostate function by primarily affecting stromal cell proliferation and angiogenesis[2,31,35,36]. The impact of estrogen on prostatic epithelium is intricate. While it has been associated with inducing hyperplasia, metaplasia, and keratinization of the epithelium, as well as neoplasia in the prostate of adult rodent models[37-39], studies have also shown the administration of estrogen-induced BPH in dogs. Moreover, the estrogenic effect has been demonstrated to induce aberrations in the prostatic epithelium in both primates and humans[39,40]. Therefore, maintaining the estrogen/testosterone (E/T) ratio is integral for maintaining normal function and homeostasis of the prostate gland[39]. Besides androgens and estrogens, progesterone, prolactin (a hypophyseal hormone), and insulin regulate prostatic function and growth[2].

Steroid action in the prostate gland is mediated through respective intracellular hormone receptors. The human prostate expresses AR, estrogen receptors α and β (ERα and ERβ), estrogen-responsive G protein-coupled receptor 30 (GPR30), progesterone receptor (PR), and glucocorticoid receptor (GR)[2]. Following puberty, the steady state phase of the prostate gland is maintained by balancing cell proliferation and cell death, a process regulated by AR signaling in both the epithelium and stromal cells of the prostate[41]. During the prenatal and postnatal differentiated stage, ERα is primarily expressed in the stroma and smooth muscle cells, while ERβ is expressed in the epithelium[42-44]. The compartmentalization of expression and the differential affinity of both ERα and β to bind to ligands and cofactors suggests the diverse functional role of estrogen within the prostate gland. Experiments in mice show that ERα-regulated transcription of cytokine genes in the mesenchyme regulates prostate differentiation and morphology during development. Meanwhile, estrogen-mediated signaling in the prostate epithelium, mediated via ERβ, has been shown to be important for epithelial function[39].

Isoforms of PR (PRA and PRB) are predominantly expressed in the stromal and smooth muscle cells of the prostate. The interaction with the prostate epithelium is crucial for PR expression in the stromal cells. Notably, PR activation has been observed to inhibit stromal expansion, which contrasts ERα regulation of stromal cells[45]. These observations suggest the coordinated activity of PR and ERα in maintaining epithelium-stromal homeostasis in the prostate gland; however, the mechanisms remain unclear. Glucocorticoids exert pleiotropic effects systemically through the GR receptors. In the prostate, GR and AR share overlapping cistromes and transcriptomic signatures. AR activation has been shown to downregulate GR expression in the prostate epithelium, indicating a critical negative feedback regulation between these two hormone receptors. Consequently, in castration conditions, GR signaling can bypass AR inhibition, promoting therapeutic resistance and prostate cancer cell survival[46].

IMMUNE LANDSCAPE IN PROSTATE GLAND

Tumor-infiltrating immune cells influence the progression of prostate cancer and its response to treatment, yet understanding the immune microenvironment crucial for normal prostate function remains limited. Recent studies utilizing bulk and single-cell sequencing techniques on normal and non-cancerous prostate tissue have identified a diverse array of leukocytes, including mononuclear phagocytes (MNPs), mast cells, natural killer (NK) cells, B cells, and tissue-resident T cells. Cross-species analysis in mice has revealed that T cells and MNPs, which persist even in prostate cancer, primarily populate the healthy prostate. Cross-analyses of single-cell RNA sequencing datasets in humans have identified six distinct classes of MNPs in the normal prostate, including monocytes, conventional dendritic cells (cDC1 and cDC2 subsets), proliferating macrophages, and various macrophage subclasses, including MAC1, MAC2, and MAC-MT[47,48].

Tissue-resident macrophages are crucial during embryonic development and in maintaining adult tissue homeostasis[49]. The diversity observed among MNPs within the prostate suggests potential tissue-specific functions. Transcriptomic analyses reveal that certain macrophage subclasses, such as MAC-MT, are exclusive to the prostate gland and exhibit heightened expression of zinc transporter genes, implicating their involvement in zinc homeostasis critical for prostatic fluid synthesis[50,51]. Remarkably, targeted depletion of macrophages using antibody targeting colony-stimulating factor 1 receptor (CSF1R) resulted in decreased zinc concentration specifically within the prostate gland, confirming the presumed role of MAC-MT macrophages in zinc regulation[47]. Furthermore, the distinct functions of dendritic cell subsets, such as cDC1 and cDC2, in antigen presentation and T-cell activation, respectively, underscore the dual importance of MNPs in both maintaining prostatic function and regulating immune responses[47,48].

Contrary to conventional theory, there is growing evidence to suggest that tissue-resident macrophages are seeded in the embryonic stage and self-maintained throughout adulthood. Transcriptomic analyses revealed that MAC-MT, with upregulated levels of zinc transporters genes, SLC39A8 and SLC30A1, might be seeded in the prostate from the prenatal stage, as it is transcriptionally similar to the yolk sac-derived macrophages[47]. The expression of the zinc transporters and metallothionein genes were also identified to be highly expressed in embryonically seeded macrophages in the murine prostate gland, further highlighting the developmental origin of these specialized immune cells. Conversely, MAC2 was suggested to be monocyte-derived and specific to the prostate gland[47-49,52]. These insights underscore the intricate interplay between immune cell populations and tissue-specific functions within the prostate, providing valuable insights into potential therapeutic targets for prostate cancer and related disorders.

HORMONAL DYSREGULATION IN AN AGING PROSTATE

Hormonal imbalances, tissue atrophy, and chronic inflammation are characteristic features of an aging prostate[53]. Additionally, with aging, there is an increased likelihood of transitioning from adult differentiated biology to repair-centric/emergent systems biology in tissues, resulting in the activation of reactive stromal response. This reactive stroma plays a crucial role in regulating epithelial proliferation and modulating the immune microenvironment. Notably, hormonal dysregulation is one of the primary contributors to the transformation of the prostate stroma into a reactive phenotype[53].

Aging men experience an upregulation of estrogen production due to declining testosterone levels, a process exacerbated by comorbidities like obesity and type 2 diabetes[53-56]. Testosterone deficiency is implicated in inducing chronic inflammation within the prostate tissue, as testosterone plays a crucial role in inhibiting the pro-inflammatory response of prostate stromal cells by activating AR and inhibiting the secretion of inflammatory cytokines and growth factors[57,58]. Additionally, testosterone protects against inflammation caused by uropathogenic bacteria like Escherichia coli by downregulating the Janus Kinases (JAK)/signal transducer and activator of transcription 1 (STAT1) signaling pathway in the prostate epithelium[59]. Indeed, chronic inflammation of the prostate gland induced by bacteria was observed to induce premalignant and malignant lesions in the prostate gland of Mongolian gerbils[60].

Obesity exacerbates hormonal imbalance by increasing aromatase activity, leading to the conversion of testosterone to estradiol, the most potent form of estrogen in men[61]. The increase in the estrogen/testosterone (E/T) ratio due to aging can lead to estrogen dominance, promoting stromal cell proliferation and fibrosis, which can accelerate clinical progression in BPH, and induce premalignant lesions in the prostate gland[62,63]. Racial disparities in prostate cancer incidence and mortality rates, particularly among African American men, have been linked to dysregulated estrogenic action on the prostate gland, with higher serum levels of estradiol observed in Non-Hispanic black men compared to Non-Hispanic white men[39,64-68]. Additionally, exposure to elevated estrogen levels during early gestation has been suggested to be a contributing factor to racial differences in prostate cancer risk, which needs to be substantiated through population-based studies. However, in vivo rodent studies support the induction of abnormalities in the prostate gland by early estrogenic exposure[37,69-71].

Multiplex profiling has revealed a significant enrichment of T and B lymphocytes in the mouse prostate gland due to aging. Specifically, a strong correlation was observed between age and enrichment of programmed cell death protein 1 positive (PD-1+) CD4+ and CD8+ T cells in the mouse prostate gland. PD-1, a cell membrane protein, plays a critical role in inhibiting both T and B-cell immune response, and is a marker for T-cell "exhaustion"[47,53,72]. Intriguingly, estrogen modulates immune cells of both myeloid and lymphoid lineages in a tissue-context-dependent manner[73]. Given the presence of both myeloid and lymphoid cells in prostate gland homeostasis[47,48], understanding the dynamic changes in the immune profile within the prostate gland in the context of testosterone deficiency and estrogenic dominance is crucial for comprehending disease development in an aging prostate.

TUMOR-STROMA INTERACTIONS

While tissue-emergent/reactive stromal responses in carcinomas are often considered secondary to epithelial changes, the limited progression of many epithelial tumors from in situ lesions despite harboring genetic abnormalities associated with malignancy raises questions about the driving molecular factors of neoplasms[74,75]. Recombinant studies in murine models have provided evidence suggesting that the stromal microenvironment is a key determinant in promoting prostate carcinogenesis[76-80]. Moreover, exogenous insults directly affecting the stroma have been identified as critical initiators of the carcinogenic process in various other solid tumors. For instance, ultraviolet radiation-induced dermal atrophy has been shown to precede keratinocyte tumors, while chemicals in cigarette smoke metabolically promote cancer by inducing autophagy and premature aging in the host stromal microenvironment in an organ such as the breast[75,81,82]. In another example, obesity-induced metabolites derived from gut microbiota induce senescence in hepatic stellate cells, which then secrete inflammatory and tumor-promoting factors that facilitate the development of hepatocellular carcinoma in mice exposed to chemical carcinogens[83]. Collectively, these instances suggest that the tissue stroma may indeed play a primary role in initiating and promoting cancer development.

Thus, the experimental evidence cited above suggests that aging or insult-driven changes of the stroma create a permissible emergent/reactive tissue or organ environment (soil) that promotes the growth of monoclonal or polyclonal tumors (field cancerization). However, based on this view, sustainable treatment or cure for cancer will be difficult to attain as long as the reactive soil persists[75]. Therefore, there is a pressing need to characterize the stromal compartment of solid tumors. One of the main limitations in characterizing reactive stroma in prostate cancer is the heterogeneity of cancer-associated fibroblasts (CAFs) that make up the TME. Stromal heterogeneity is partially explained by the fact that CAFs can be derived from the activation of tissue-resident fibroblasts, mesenchymal stem cells, vimentin-positive periacinar cells, circulating bone marrow-derived precursors, vessel-associated pericytes, and endothelial cells[10,11]. Spatial transcriptomic analysis of radical prostatectomy-derived tissue, in addition to stromal cell lineages identified from single-cell sequencing analyses of mouse prostate stroma, suggests the presence of reactive stromal cells with different transcriptional programs and functions within the prostate cancer TME[84,85]. Additionally, the phenotypic plasticity of the activated stromal cells further underscores the dynamic nature of the reactive stroma[11]. Hence, characterizing a moving target such as TME to understand tumorigenesis, development, and progression becomes a challenging endeavor.

STROMAL RESPONSE IN PROSTATE CANCER

The coordinated host emergent response to tissue injury involves the collective action of cells that make up the connective tissue/stroma and the extracellular matrix (ECM) products. The normal reactive stromal response to injury is self-limited and regulated spatially and temporally to re-establish tissue integrity and reset homeostasis. The mechanisms underlying reactive/emergent stromal response include the release of inflammatory and growth signals, basement membrane dysfunction initiating cell-to-matrix and intercellular interactions regulating cell proliferation, migration, and differentiation of both stromal and epithelial cells, fibroplasia, angiogenesis, ECM remodeling, and wound contraction [Figure 1]. However, the persistence of this repair process resulting in a chronic, non-healing wound and fibrosis, can affect any tissue and organ system in the human body[86-88].

In 1863, Rudolf Virchow first recognized the association between wound healing phases in tissue and tumorigenesis. Clinical similarities suggested shared common cellular and molecular signatures between the two conditions. This insight led Scottish pathologist Dr. Alexander Haddow to deduce cancers as wounds that overheal[89,90]. However, clinical evidence indicated that in cancer, reactive stroma is not self-limited and is tumor-promoting, which led Dr. Harold Dvorak to postulate that "tumors are wounds that do not heal"[86]. Owing to Paget’s "seed and soil hypothesis", reactive stroma has emerged as essential soil regulating multiple aspects of tumorigenesis, including initiation, development, progression to metastases, and most importantly, development of therapeutic resistance[91].

Reactive stroma is heterogeneous in its makeup, exhibiting both organ- and tumor-specific characteristics. The prevalence and abundance of reactive stroma serve as disease-defining factors and are associated with poor prognosis in several solid tumors, including colon carcinoma, head and neck cancer, HER2-negative early breast cancer, squamous cell carcinoma, and rectal cancer[20,92-96]. In prostate cancer, reactive stroma co-evolves with tumor development, and its relative abundance is quantified as reactive stromal grade (RSG). An RSG of 3 represents when more than 50% of the prostate tumor area is composed of reactive stroma, and the latter is associated with earlier biochemical recurrence and worse prognosis[97,98]. Reactive stroma in prostate cancer is composed of CAFs that can transdifferentiate into cancer-associated myofibroblasts (myCAFs) or inflammatory fibroblasts (iCAFs), with an expanded and modified ECM with collagen deposition, dense microvessels, and immune infiltrates [Figure 1][11,17,99]. Tissue recombinant experiments demonstrated that prostate cancer-derived CAFs promote tumor growth in vivo while normal fibroblasts inhibit the process, thus confirming the critical nature of reactive stroma in prostate tumor growth and development[100-102].

MOLECULAR FEATURES OF REACTIVE STROMA

TGF-β signaling

Cytokines, such as TGF-β, play a crucial role in regulating cell fate and reactive stromal response. TGF-β induces the differentiation of stromal cells into vimentin and smooth muscle alpha-actin-positive myCAFs, thereby initiating a wound repair-like reactive stroma. Concurrently, TGF-β also modulates the composition of the ECM by inducing the expression of collagen 1 and tenascin-C in stromal cells[10,17]. TGF-β is also critical in modulating the immune reaction. It can suppress interleukin 2 (IL-2) synthesis and T-cell proliferation, as well as regulate the differentiation of both CD4+ T cells and regulatory T cells (Tregs). Cytokines such as IL-10, IL-4, and TGF-β secreted by reactive stromal cells and immune-suppressive cells like Tregs can increase the polarization of M2 macrophages. M2 macrophages have been shown to enhance angiogenesis, tissue remodeling, and modulate the immune microenvironment by expressing human leukocyte antigen (HLA-DR) and programmed death-ligand 1 (PD-L1), resulting in the suppression of the immune system[103-105].

Although the expression of TGF-β increases from prostatic intraepithelial neoplasia (PIN) to the development of prostate cancer lesions[10,17], the biological activity of TGF-β within TME is not cancer-specific. TGF-β is a fundamental regulator of different cellular processes in adult differentiated biology[106]. Thus, the biological activity of TGF-β activity is essentially indistinguishable whether in a wound repair, tissue fibrosis, or TME. TGF-β, secreted by both epithelial cells and fibroblasts, is critical for coordinating tissue repair and homeostasis through its context-dependent pleiotropic functions. For example, TGF-β induces cytostasis in non-transformed epithelial cells, while in endothelial and mesenchymal cells, it stimulates proliferation. Additionally, TFG-β can both induce and suppress apoptosis, suggesting that other critical signaling inputs mediate TGF-β-induced cellular survival and cell death[107]. Epithelial-to-mesenchymal transition (EMT) is an important cellular event common in wound repair, fibrosis, and cancer. TGF-β-induced signaling, either independently or in coordination with critical mediators of tissue homeostasis such as Wnt and NOTCH, induces EMT by repressing inter-epithelial adhesion and junctional complexes while upregulating mesenchymal markers including ECM remodeling enzymes like matrix metalloproteinases (MMPs), facilitating cell migration[108]. Apart from TGF-β, growth factors such as FGFs and PDGF, which are critical for wound healing and tissue remodeling, also regulate reactive stromal biology in prostate cancer[109,110].

Remodeled ECM

ECM remodeling serves as another critical regulator of tissue biology, integrity, and, most importantly, maintaining homeostasis. Beyond providing physical support, the ECM plays a pivotal role in cell adhesion, migration, initiating angiogenesis, tissue development, and repair[111]. The cells of the reactive stroma actively overexpress ECM proteins (collagens, elastin, fibronectin, tenascin-C, and hyaluronic acid), MMPs, fibroblast activation protein (FAP), and lysyl oxidases[17,111-113]. MMPs, in particular, play a critical role in remodeling the ECM and inducing modifications that influence its topographical and mechanical properties. In healthy tissue, ECM remodeling is a tightly regulated process that involves the counterbalance of ECM synthesis with the activity of MMPs, MMP inhibitors, and lysyl oxidases[114].

Damage to the ECM due to tissue injury initiates an emergent response referred to as a "wound healing cascade" to repair damaged tissue and reset tissue homeostasis[111]. In cancer, ECM is damaged and exposure to chronic remodel signals generated from both the cancer cells and reactive stromal cells results in a chronic wound-healing cascade. Consequently, the dysregulated ECM remodeling and turnover results in the development of fibrotic tissue (desmoplasia) with enhanced stiffening around the tumors. This ECM alteration affects every aspect of tumor biology, including the regulation of proliferation, differentiation, gene expression, cell adhesion, migration, invasion, etc.[111]. Beyond its regulatory control, ECM also influences immunogenicity, oxygenation, and the response of cancer to treatments. Thus, a reactive stroma composed of extensively remodeled ECM is correlated with poor prognosis in cancer[111,115].

Activated angiogenic niche

Cells within the vasculature network, primarily endothelial cells (EC), pericytes, and vascular smooth muscle cells, constitute additional critical components of the tissue stroma and are essential regulators of prostate homeostasis[10]. In healthy tissue, the non-angiogenic EC interacts with a complex basement membrane composed of collagen IV, laminin, perlecan (heparan sulfate proteoglycan), and entactin/nidogen via integrin[116]. However, in response to wounding and cancer, the reactive stromal cells (myofibroblasts or CAFs) secrete ECM such as collagen I and IV, fibronectin, secreted protein acidic and rich in cysteine (SPARC), tenascin, heparan sulfate proteoglycans, connective tissue growth factor, and VEGF. On interaction with these reactive stromal products, the quiescent EC transitions into an activated status/angiogenic switch, resulting in the formation of phenotypically distinct blood vessels with aberrant branching and enhanced leakiness[10,116]. Hypoxia is another critical feature common to both tissue injury and TME. Hypoxia-induced activation of hypoxia-inducible factors (HIFs) in both epithelial and stromal cells results in the secretion of proangiogenic factors that modulate vessel maturation. Thus, the new growth of vascular network in response to reactive stromal response in the TME is critical in regulating the proliferation, growth, and progression of solid tumors[117-119].

In addition to providing nutritional support and oxygen supply, activated EC express chemokines and adhesion molecules, facilitating the recruitment of leukocytes, monocytes, and neutrophils[120,121]. Pericytes, a class of tissue-resident mesenchymal stem cells, play a critical role in the stabilization of neovessels and regulating MMP activity[10,99,116]. In both wound repair and cancer, pericytes deviate from the EC and vascular basement membrane, undergoing a phenotypic transformation that regulates angiogenesis, inflammation, fibrosis, tissue regeneration, and re-epithelialization[122,123]. All these functional aspects of pericytes in an emergent wound repair scenario are critical in establishing a pre-metastatic niche, which is critical for tumor growth and progression[124].

STROMAL MEMORY

The concept of cellular "memory" can be defined as when cells maintain an altered phenotypic or functional state proceeding with an initial environmental stimuli/insult. Myeloid lineage cells like monocytes, NK cells, macrophages, and neutrophils exhibit innate immune memory, a manifestation of cellular memory[125,126]. Research over the past two decades has demonstrated that these cells exhibit protective or cross-protective mechanisms against recurring infections through heightened activation of the innate immune response. This heightened response is driven by pattern recognition receptors (PRRs) on myeloid cells, allowing recognition of pathogen-associated molecular patterns (PAMPs). Activation of PRRs by PAMPs triggers the expression of genes involved in inflammatory and immune responses[127-129].

Fibroblast memory

Emerging evidence suggests that non-immune cells also possess a memory of past insults such as inflammation, enabling them to mount rapid responses to emergent situations like injury or infection. The biological mechanisms regulating cellular memory are multifaceted, involving various processes such as alterations in chromosomal accessibility due to epigenetic modifications, increase in expression of activation receptors, and priming of cellular signaling networks[121,130-133].

Naik et al. were the first to discover that epithelial stem cells exposed to inflammation retain cellular memory, leading to enhanced repair responses to future tissue-related injuries[121,134]. This phenomenon extends to fibroblasts, which develop an inflammatory memory upon exposure to exogenous challenges such as lipopolysaccharide (LPS) or endogenous inflammatory signals like tumor necrosis factor α (TNF-α)[121]. For instance, human gingival fibroblasts pretreated with LPS showed no tolerance but maintained cytokine and chemokine expression after secondary LPS treatment[135]. Similarly, in conditions like rheumatoid arthritis, fibroblasts such as synoviocytes exhibit gene-specific priming by altering chromatin following chronic exposure to inflammatory signals like TNF-α, leading to enhanced and prolonged chemokines and cytokine production upon subsequent interferon (γ) stimulation[132,136]. Klein et al. have also observed that LPS primes synovial fibroblasts to sustain inflammatory responses by changing the epigenetic configuration at gene promoters regulating LPS-induced cellular responses[131].

In patients with tendinopathy, stromal fibroblast activation markers such as podoplanin and vascular cell adhesion molecule (VCAM-1) are notably elevated compared to healthy tendon tissues. This elevation persists even after the gradual decline in inflammatory gene signatures following the removal of stimuli like IL-1β, suggesting that activated fibroblast memory maintains a persistent activated state rather than sustaining inflammatory responses[130]. Additionally, in rheumatoid arthritis, sustained synovial inflammation is also attributed to persistent activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling induced by TNF-α, facilitated by upregulation of TNF receptors and proximal signaling components, and downregulation of negative feedback inhibitors involved in the homeostatic balance of the NF-κB signaling pathway[136]. Epigenetic modifications are also known to regulate epithelial stem cell memory[121,131,132,134].

Endothelial cell memory

Similar to fibroblasts, EC is also activated in response to inflammation. EC activation can be distinguished as a "delayed (type II)" or "immediate (type I)" response. In the delayed response, upon activation, EC expresses chemokines, VCAM-1, intercellular adhesion molecule (ICAM-1), and E-selectin several hours post-stimulus due to a requirement for de novo transcription and translation[137,138]. In contrast, in the type I response, there is no delay following stimulation and response due to the preformation of adhesion molecules and chemoattractant resulting from a preceding inflammatory stimulus. This suggests EC memory[138-141]. Studies have demonstrated that EC exposed to homocysteine, an independent risk factor for developing atherosclerosis, has an augmented response to inflammatory mediators such as LPS and thrombin[142]. Additionally, recent studies have also shown that EC stores a metabolic memory of an earlier transient hyperglycemia in the vasculature in diabetic patients, resulting in epigenetic changes, cardiovascular complications, chronic inflammation, and oxidative stress in later stages[143-145].

As integral components of the tissue structure, the ability of stromal cells to adapt to environmental stimuli while retaining memory of past exposures is essential for maintaining tissue homeostasis[125,146,147]. Various environmental stimuli, such as injury or infection, can trigger the innate cellular memory in stromal cells, leading to an activated phenotypic and functionally emergent state through different molecular mechanisms, as described. This emergent microenvironment state of tissues in diseases like cancer can have a detrimental impact on disease initiation and development, progression, and response to treatments[21,125,148-150]. For instance, chronic inflammation of organs due to injury-causing agents or infections is known to induce cancers, such as esophageal, lung, gastric, and colon cancer, which are often metastatic, treatment-resistant, and lethal[151-158]. Therefore, targeting specific stromal memories involved in maintaining a reactive/emergent stromal response emerges as a promising therapeutic strategy to mitigate the detrimental effects of microenvironment priming and enhance treatment efficacy in cancer patients[21,125,159].

IMMUNE REGULATION IN CANCER

Traditionally, cancer research has focused on the intrinsic biology of cancer cells to identify potential molecular determinants crucial for tumor growth, development, and progression. However, there has been a recent shift in attention toward the role of non-cancerous cellular components in the TME, particularly immune cells, in controlling tumor growth and development. This shift has been clinically validated and garnered attention because of its potential to be curative in subsets of cancer patients[150]. Consequently, there has been an increase in research efforts aimed at understanding the mechanisms regulating the reactivity of immune cells toward various types of tumors. This shift in focus reflects a growing recognition of the intricate interplay between cancer cells and the TME, highlighting the importance of comprehensively understanding the latter for the development of effective cancer therapies.

Paul Ehrlich's hypothesis on immune cells suppressing carcinoma development led to the "immune surveillance hypothesis" later proposed by Burnet and Thomas[160]. While the tumor-specific immune response was validated in inbred mouse strains, discordant results from immune-deficient mouse models initially cast doubt on the concept[161]. However, the development of defined immune-deficient models and epidemiological data from human studies reaffirmed the relevance of cancer immune surveillance, leading to the broader concept of "cancer-immuno-editing". This concept recognizes the dual role of host-protecting and tumor-sculpting properties of the immune system. Cancer-immuno-editing involves three stages: tumor elimination by the immune system (immune surveillance), a phase of equilibrium where tumor variants resistant to surveillance are selected (immune-sculpting), and the emergence of overt tumors in an immunocompetent host (immune-escape)[161-167].

Role of stroma in immunomodulation of the TME

The stroma plays a pivotal role in immunomodulation, posing a significant challenge to immunotherapies in various solid tumors like pancreatic duct adenocarcinoma (PDAC), non-small cell lung cancer, ovarian cancer, hepatocellular carcinoma, and prostate cancer[23,168-172]. However, targeting the stroma to enhance immunotherapy efficacy and hinder tumor progression has been largely overlooked. Understanding the complex interplay between tumor-stroma-immune components is crucial for developing innovative therapies to modulate the TME and improve targeted cancer treatments[21,161].

Stromal cells significantly contribute to cancer-immuno-editing by modulating the immune system through the secretion of various chemical messengers, such as chemokines, cytokines, and prostaglandins, as well as the ECM. In PDAC, elevated levels of the chemokine-chemokine (C-X-C motif) ligand 10 (CXCL10), positively correlated with high stromal content, are associated with decreased median overall survival in patients. CXCL10 expression is linked to the presence of Tregs, which exerts immunosuppressive effects, compromising immune surveillance against cancer[21,173,174]. Additionally, the immune regulatory chemokine CXCL5 secreted by tumor-associated macrophages (TAMs), CAFs, EC, and cancer cells themselves plays a crucial role in recruiting neutrophils to the TME. Neutrophils, in turn, modulate the TME, promoting tumor growth and progression, and induce anti-inflammatory M2 macrophage polarization, impairing immune surveillance. High expression of CXCL5 is associated with poor patient survival in various cancers, including renal, pancreatic, liver, and cervical cancer[21,175-177].

In solid tumors, myCAF-expressing FAP exhibits immunosuppressive properties by secreting large amounts of stromal cell-derived factor-1 (SDF-1), hindering T-cell-tumor interactions and attracting Tregs. Ablation of FAP+ stromal cells leads to hypoxia-induced cancer cell death mediated by interferon-γ and TNF-α[178-181]. Additionally, CAF-secreted TGF-β inhibits host immune surveillance by impairing dendritic cell, M1 macrophage, NK cells, and CD8+ T-cell function, while promoting Treg and Th17 cell differentiation and suppressing B cell proliferation and IgA secretion[182,183]. Moreover, TGF-β restricts T-cell infiltration, diminishing tumor response to PD-L1 blockade[184].

TGF-β induces ECM remodeling, while its suppression of ECM-modulating proteins like MMP-1, -8, and -13 results in the formation of fibrotic and desmoplastic ECM matrix, which is associated with cancer recurrence and chemoresistance[185-187]. Desmoplasia impedes T-cell recruitment into tumor nests, causing T-cell accumulation in peri-tumoral regions and promoting immune escape. Additionally, within these peri-tumoral regions, T cells are exposed to paracrine signals, resulting in their suppression[23]. The matricellular protein periostin (POSTN) is highly expressed by both tumor and stromal cells. Its elevated expression is associated with poor prognosis in various cancers, including prostate, lung, pancreatic, ovarian, breast, colorectal, hepatocellular, bladder, and osteosarcoma[188]. POSTN promotes PD-1 expression in TAMs via integrin-ILK-NF-κB signaling. PD-1-expressing TAMs were observed to induce PD-L1 expression in colorectal cancer cells, promoting immune escape[189]. Collectively, the reactive stromal response plays a critical role in tumor development, progression, and modulation of the immune landscape in TME [Figure 2].

The reactive stroma response regulates the immune landscape in prostate cancer

Figure 2. Emergent stromal response regulate immunosuppressive landscape in solid tumors. (A) The reciprocal interactions between tumor cells (T) and expanding CAFs within the reactive TME results in the secretion of TGF-β and various chemokines (CXCL) facilitating the recruitment and regulation of Tregs. (B) Treg secreted cytokines (IL-4, IL-10 and TGF-β) trigger polarization of M1 macrophages to the M2 phenotype. (C) PD-L1-expressing M2 macrophages induce T-cell exhaustion. (D) TGF-β in the TME derived from different cellular sources can modulate the extracellular matrix (ECM) composition. Both TGF-β and the modified ECM impedes tumor-infiltrating lymphocytes (TIL) both molecularly and mechanically. (E) The reactive stroma induces angiogenesis, further supporting tumor growth and survival. (F) Matricellular protein-periostin (POSTN), expressed during reactive stromal response attract TAMs. TAMs facilitate immune evasion in tumor cells by inducing the expression of PD-L1. TAMs also recruit neutrophils, which induces an immunosuppressive TME by causing M2 polarization. Both TAM and neutrophils also induce therapeutic resistance.

THE IMMUNE LANDSCAPE IN PROSTATE CANCER

In the past decade, the understanding of the immune landscape in cancer has evolved significantly. Studies have utilized cell surface markers to identify various immune cell populations within the TME, including T cells, B cells, NK cells, macrophages, monocytes, and granulocytes. Solid tumors are commonly classified as having "hot" or "cold" immune microenvironments based on the presence or absence of these immune cell populations within the tumor margins[167,190,191]. Prostate tumors are typically classified as having a "cold" TME, characterized by elevated PD-L1 expression and lower levels of tumor-infiltrating immune cells, like CD3+ T cells, CD20+ B cells, and CD68+ macrophages compared to BPH[192,193]. In addition to elevated levels of Tregs and myeloid-derived suppressor cells (MDSCs), contributing to an immunosuppressive microenvironment in prostate cancer[167,194], the disease also exhibits reduced tumor antigens due to its low tumor mutational burden[195]. Additionally, AR signaling suppresses major histocompatibility complex 1 (MHC1) expression and T-cell response, further complicating the development of immunotherapy to target prostate cancer[167,196-198].

In primary prostate cancer, both the cancerous epithelium and stromal cells express inflammatory factors like TNF-α and IL-6, which induce reactive oxygen species, leading to inflammation, immunosuppression, and tissue damage[199]. TNF-α and IL-6 promote treatment-resistant/castration-resistant prostate cancer (CRPC) by affecting stromal and prostate cancer cells[199,200]. IL-6 specifically, mediated by bone morphogenic protein (BMP) and CD105, induces androgen receptor splice variant 7 (AR-V7) expression in prostate cancer cells and fibroblasts, a key mechanism in CRPC progression. IL-6-mediated AR-V7 expression in fibroblasts induced resistance to anti-AR inhibitors in prostate cancer cells. However, neutralizing CD105 downregulated AR-V7 in both prostate cancer cells and fibroblasts, resensitizing the cancer cells to these inhibitors[201]. Additionally, paracrine interactions between epithelial and stromal cells stimulate prostate stromal cells to secrete chemokines such as CXCL-1, CXCL-2, CXCL-3, and IL-8 (CXCL-8)[199], which recruit leukocytes like neutrophils, macrophages, monocytes, and MDSCs into the microenvironment via C-X-C chemokine receptor type 2 (CXCR2) activation[202-204]. In a murine prostate cancer model, CXCR2 knockout or antagonist administration reduced tumor growth and shifted tumor-associated macrophages toward a pro-inflammatory M1 phenotype[205]. Elevated neutrophil count correlates with worse overall survival in localized prostate cancer[206], while elevated IL-8 levels in prostate cancer patients with metastatic disease starting on androgen deprivation therapy (ADT) were associated with shorter progression time to castration resistance and overall survival[207]. Therefore, understanding the mechanisms governing the inflammatory and immunosuppressive TME from localized disease to metastasis is essential for developing effective immunotherapies for advanced prostate cancer.

To characterize the immune microenvironment in solid tumors like prostate cancer, numerous scientific groups and biotechnological companies have devoted considerable efforts to developing tools for genetic, transcriptomic, metabolic, and proteomic-based profiling of the immune landscape. These approaches encompass serum, spatial, and single-cell-based techniques[47,208-212]. Notably, there have been significant strides in developing immune-related gene signatures to elucidate key immune cell components within prostate cancer, such as macrophage-based gene signatures[210], metabolic syndrome-based index scores[213], immune subtyping[214,215], immune-based risk scoring[216], and long non-coding RNA signatures[217]. These advancements significantly contribute to the capacity to develop biomarkers for diagnostic and prognostic purposes, enhancing the ability to assess the risk, progression, and sensitivity to immunotherapies in prostate cancer.

Research efforts aimed at transforming "cold" immune microenvironments into "hot" ones in solid tumors, including prostate cancer, are underway to enhance the efficacy of immune checkpoint inhibitors (ICIs) like PD-1/programmed death-ligand 1 (PD-L1) or cytotoxic T-lymphocyte–associated antigen 4 (CTLA4) inhibitors[197]. However, a significant challenge in this strategy, particularly in prostate cancer, is the reactive stromal response, which can molecularly and mechanically shield tumor cells from the antitumor immune response. Additionally, the dysregulation of critical enzymes involved in cellular energetics within tumors, including prostate cancer, is emerging as a hallmark feature associated with tumor evasion, though the mechanisms are complex[218,219]. Therefore, comprehending the dynamic intercellular crosstalk between tumor-stroma-immune cells and its subsequent modulation of the TME, which can exclude T-cell infiltration or inhibit T-cell function, may be crucial for achieving sustainable efficacy with immunotherapies, including ICIs, in prostate cancer[23,220].

CURRENT STATE OF PROSTATE CANCER IMMUNOTHERAPIES

The current landscape of immune-based therapeutics for prostate cancer encompasses several approaches, including cancer vaccines, ICIs, adoptive cell therapies, targeted antibodies, and oncolytic viral therapy[196,221-225]. Cancer vaccines aim to stimulate the patient’s immune system, eliciting a response against tumor-specific or tumor-associated antigens (TAA). One notable example is Sipuleucel-T, an FDA-approved autologous vaccine that utilizes dendritic cells (DC) stimulated to target prostatic acid phosphatase (PAP), a protein highly expressed in prostate cancer. Clinical data show evidence of improved median survival and prolonger overall survival among men with metastatic castration-resistant prostate cancer (mCRPC) treated with Sipuleucel-T compared to those treated with mainline treatments (anti-hormone treatment ± chemotherapy). While Sipuleucel-T treatment has demonstrated a broad and durable systemic immune response, clinical data suggest that the treatment provides greater benefits to patients with a lower disease burden. This observation may be attributed to the existence of a robust immune system at the initial stages of cancer development, in contrast to more advanced disease states. In addition, treatment with Sipuleucel-T did not significantly affect mCRPC disease progression. This may be due to the delayed onset of antitumor response by Sipuleucel-T, and therefore, a timeline of diagnosis of mCRPC may play an important role in determining the maximum possible benefit from Sipuleucel-T treatment[226,227]. Additional mechanistic underpinnings that resulted in the limited efficacy of DC vaccine for clinical management of prostate cancer are described further in this review. However, several vaccine targets are currently under evaluation against prostate cancer, including oncofetal antigen-5T4, carcinoembryonic antigen (CEA), PSA, prostate-specific membrane antigen (PSMA), survivin, tumor-associated antigens (TAA), and personalized neoantigens. These selected proteins serve as targets due to their high expression levels in cancer cells compared to their normal counterparts[228].

Other prominent strategies in prostate cancer immunotherapy involve the use of immune modulators to disrupt immune checkpoints, such as PD-1/PD-L1 and CTLA-4, exploited by cancer cells to evade immune detection and responses, often leading to T-cell exhaustion[196]. In addition to these well-established targets, ongoing clinical investigations in other urological cancer types, such as bladder cancer, explore immune modulators targeting the immunosuppressive activity of CD73, indoleamine 2,3-dioxygenase (IDO), and lymphocyte activation gene 3 (LAG3)[229-232] (Clinical Trial Registration Numbers NCT03454451, NCT05843448, NCT04586244). Immunomodulation treatments also include the activation of co-stimulatory pathways to promote or enhance T-cell functions by downregulating immunosuppressive components like Tregs in the TME. Key targets in this category include inducible co-stimulator (ICOS), OX40, Toll-like receptors (TLRs), CD137, and IL-2/IL-2R[233-236].

Adoptive or cell-based immunotherapy represents another autologous approach, where the patient’s immune cells, such as T cells, are isolated, expanded in vitro, and modified with chimeric antigen receptors (CARs) that can specifically target antigens expressed by tumor cells, thereby eliminating them. Adoptive immunotherapy targets currently under evaluation for prostate cancer include prostate stem cell antigen (PSCA) and PSMA[237]. Beyond T cells, both NK and tumor-infiltrating lymphocytes (TILs) can also be enhanced and reintroduced into patients[238,239]. Monoclonal antibodies constitute another class of treatments developed to block specific cell membrane receptors from binding to its target ligand, thereby impeding its functional impact on cancer growth and proliferation. Commonly targeted membrane receptors in prostate cancer include delta-like proteins (DLL), Notch, human epidermal growth factor receptor 2 (HER2), and tumor-associated calcium signal transducer 2 (TROP2)[240-242]. Antibodies can also be modified to carry cytotoxic payloads, specifically chemotherapeutics, for their active delivery to tumors[243]. Bi-specific T-cell-engaging antibodies or BiTEs bind to cancer cells and T cells, activating the latter[244]. Oncolytic viral therapy involves the use of different DNA (Adenovirus and Herpes simplex virus) and RNA (Reovirus) viruses, often modified to infect tumor cells and induce cell death. This approach can elicit an immune response that further aids in the elimination of both localized and metastatic tumors[245].

While appealing, the immunosuppressive and "cold" TME in prostate cancer poses a significant challenge for the immunotherapy strategies described. Therefore, emerging therapeutic approaches aim to target both cancer cells and the TME. One strategy involves directly targeting stromal markers upregulated in the TME, such as FAP, which is associated with poor prognosis in various solid tumors. Targeting FAP-expressing CAFs using CAR-T therapy shows promise in improving tumor-targeted cytotoxicity of CAR-T cells targeting solid tumors. In summary, therapies targeting both cancer cells and the TME hold the potential for effectively treating prostate cancer and improving patient outcomes. Table 1 provides a summary of various immunotherapies currently under evaluation in clinical trials, while Table 2 outlines potential therapies for targeting tumor stroma[20,245,263].

Table 1

Selected clinical trials of immunotherapies targeting cancer

TypeTreatmentCombinationTargetTumor typePhaseStatusIdentifier
Peptide vaccineNY-ESO-1 ProteinCpG 7909NY-ESO-1Adv. PCaICompleteNCT00292045
UV1GM-CSFhTERTCSPC, mPCI/IIUnknown statusNCT01784913
mAbDenosumabN/ARANKLnon-metastatic CRPCIIICompleteNCT00286091
BisphosphonateZoledronic acidN/A
Osteogenic nichemCRPC and mBCIVRecruitingNCT04549207
InhibitorsIbrutinibN/ABTK, MMP-2, MMP-9PCaIICompleteNCT02643667
IbrutinibTrastuzumabBTK, MMP-2, MMP-9HER2+ BCI/IIOngoingNCT03379428
Selected clinical trials of macrophage immunotherapies
M2/M1
reprogramming
LY3022855N/AM-CSFRmCRPC, mBCICompleteNCT02265536
MCS110Carboplatin, gemcitabineM-CSFTNBCIICompleteNCT02435680
IPI-549Tecentriq, abraxane, bevacizumabPI3K-γBC, renal cell carcinomaIIOngoingNCT03961698
852AN/ATLR7BC, ovarian, endometrial, and cervical cancersIICompleteNCT00319748
ImiquimodAbraxaneTLR7Adv. BCIICompleteNCT00821964
CP-870,893N/ACD40Adv. solid tumorsICompleteNCT02225002
AntibodiesCP-870,893Paclitxel, carboplatinCD40Solid tumorsICompleteNCT00607048
Hu5F9-G4CetuximabCD47/SIRPaSolid tumors, Adv. CCI/IICompleteNCT02953782
PLX3397EribulinCSF-1RBCI/IICompleteNCT01596751
CAR-MCT-0508N/AHER2HER2+ solid tumors, including PCaIRecruitingNCT04660929
CytokinesGM-CSFCarboplatin, cabazitaxelHSCsmNEPC, mPCIIRecruitingNCT04709276
ProscaVax
(GM-CSF, PSA, IL-2)
N/APSAPCaIIUnknown statusNCT03579654
InhibitorsCabiralizumabPaclitaxel, carboplatin, nivolumabCSF-1RTNBCI/IIOngoingNCT04331067
DaratumumabN/ACSF-1RPCaIOngoingNCT03177460
CarlumabN/ACCL2PCaIICompleteNCT00992186
AZD-5069EnzalutamideCXCR2mCRPCI/IITerminatedNCT03177187
Dendritic cell therapies for PCa
DC VaccineSipuleucel-TN/APAPmCRPCIIICompleteNCT00065442
Stapuldencel-TDocetaxel, prednisonePAPmCRPCIIICompleteNCT02111577
With tumor mRNAN/AhTERT, survivinmCRPCI/IIOngoingNCT01197625
Table 2

Potential immunotherapies targeting the TME

TME targetMolecular targetImmunotherapiesPotential desired effectsReferences
Fibroblasts
FAPFAP-CAR-T; FAP BiTEsDestroy CAFs to disrupt tumor-stroma interactions and suppress tumor growth[246-249]
SDF-1/CXCL12CDXR4 antagonist (Plerixafor)Disrupt SDF-1 signaling to reduce tumor growth and metastasis[250]
HGF/c-METmAb or nAbInhibit tumor growth and metastasis[251]
EndotheliumVEGFImmunomodulatory agents (thalidomide; lenalidomide)Inhibit angiogenesis and cancer-stroma adherence, and stimulate the immune system[252,253]
Bevacizumab; ramucirumabInhibit angiogenesis to reduce tumor blood supply[254,255]
PDGFOlaratumabInhibit stromal cell recruitment and activation to disrupt angiogenesis, stromal support, and bone metastasis[256]
ECMMMPsMMP inhibitorsInhibit ECM remodeling to reduce tumor cell migration
[257]
CollagenCollagenaseReduce ECM stiffness and density to improve drug and immune cell infiltration
TNCmIL12-R6N mAbAntitumor activity[258]
CTGFmAbModulate the TME to reduce fibrosis and enhance the efficacy of other treatments[259]
IntegrinsmAbDisrupt cell-ECM interactions, inhibiting tumor cell migration[260]
FN1mAbInhibit cell proliferation and migration[261]
TGF-βnAbReduce immunosuppression and increase immune cell infiltration in the TME[262]

MACROPHAGE THERAPY FOR METASTATIC CASTRATION RESISTANCE PROSTATE CANCER

Immunotherapy exhibits limited efficacy in advanced prostate cancer patients with mCRPC, mainly due to the immunosuppressive TME. Targeting TAMs, the predominant immunosuppressive cells in prostate cancer, presents a promising therapeutic avenue for mCRPC treatment[264]. Macrophages, once viewed as phagocytic sentinels, now demonstrate diverse roles in maintaining tissue homeostasis. Tissue-resident macrophages oversee the microenvironment, ensuring tissue integrity, facilitating cellular communication, and regulating immunological balance[265]. Conversely, monocyte-derived macrophages, recruited during inflammation, adopt either the pro-inflammatory "M1" or anti-inflammatory/reparative "M2" phenotypes, each characterized by distinct gene expression and metabolic pathways. "M1" macrophages rely on glycolysis, producing inflammatory cytokines like IL-1β, IL-12, TNF-α, and reactive oxygen species, while "M2" macrophages employ oxidative phosphorylation, secreting molecules such as arginase-1 and TGF-β[266-268].

Despite the simplicity of the "M1/M2" dichotomy, current single-cell transcriptomic data suggest a more complex landscape, acknowledging the high degree of macrophage plasticity and tissue-specificity[269]. This complexity has propelled macrophage reprogramming to the forefront as a promising therapeutic. Macrophages' dynamic transition between "M1" and "M2" states in response to environmental cues has become a focal point in disease treatment strategies, particularly by directing them toward an "M1" phenotype to initiate inflammation and restore homeostasis.

In mCRPC, TAMs play dual roles: facilitate tumor progression, and induce immunosuppression within the TME[270]. Research also underscores TAMs' significant function in fostering resistance to anti-androgen therapies. For instance, macrophages can induce ECM remodeling, reminiscent of wound healing processes. Such macrophage-mediated ECM modifications correlate with anti-androgen resistance, particularly through the activation of fibronectin-1 (FN1)-integrin alpha 5 (ITGA5)-tyrosine kinase Src signaling cascade, induced by the cytokine Activin-A[271]. Reprogramming these TAMs to an M1-like state may disrupt these pro-tumorigenic activities. Transitioned M1 macrophages could potentially reverse the immunosuppressive TME, attenuate ECM-mediated drug resistance, and amplify the efficacy of current therapies[272].

Upregulated Src kinase activity in prostate cancer bone metastases, mediated by the Activin-A Receptor as well, is associated with macrophage density and several ECM-receptor pathways[273]. Targeting this activity with the specific Src inhibitor eCF506 has shown promise in blocking enzalutamide resistance, emphasizing its therapeutic potential in mCRPC management[271]. Furthermore, strategies that modulate TAMs can engender a more antitumoral phenotype[274]. ICIs can induce M1 macrophage polarization, and the depletion of Treg cells - protectors of the tumor-friendly milieu - by anti-CTLA-4 antibodies is contingent upon macrophage-mediated actions[275]. These insights affirm the premise that macrophage reprogramming could play a crucial role in enhancing the impact of immunotherapies in mCRPC.

An alternate, emerging approach is through chimeric antigen receptor macrophages (CAR-M), which leverages the natural tumor-homing ability of myeloid cells. This therapy has shown potential advantages in infiltrating solid tumors and can release pro-inflammatory cytokines to improve the TME[276]. Furthermore, the combination of CAR-M with CAR-T cells has demonstrated synergistic action against cancer cells, exceeding the effects of either therapy alone. This synergy suggests that CAR-M and CAR-T can complement each other, enhancing tumor responses. Despite its promising antitumor activity demonstrated in animal experiments, CAR-M therapy faces several challenges that need to be addressed. These include optimizing the CAR structure by incorporating tandem activation domains or pro-inflammatory cytokines to enhance its effectiveness and safety for clinical application[277].

Thus, the intricate relationship between macrophages, ECM components, and other immune cells within the TME underscores the potential of therapeutic interventions that manipulate these interactions, particularly through M1/M2 reprogramming and CAR-M therapy. Such a strategy could potentially disrupt critical resistance mechanisms and forge a more robust immune response against tumors in their advanced stages.

THE PITFALLS OF DENDRITIC CELL THERAPY IN PROSTATE CANCER

DC therapy has encountered significant challenges, primarily attributed to the inconsistency in clinical responses[278]. This is compounded by the fact that no DC therapies have received FDA approval since Sipuleucel-T in 2010. While certain patients exhibit enhanced immune reactions and positive clinical outcomes, such as reduced PSMA levels and tumor regression, these effects are not universally observed[279,280]. This variability raises questions about the therapy's reliability, making its therapeutic value uncertain.

The effectiveness of DC therapy hinges on the selection of appropriate TAAs and the successful maturation of the DC themselves. Common TAAs in prostate cancer - such as PSA, PSMA, PAP, and PSCA - show variable expression across different tumors, affecting the efficacy of the therapy[281,282]. Additionally, the immunogenicity of these antigens may not always be sufficient to induce a strong immune response[283]. Compounding these issues are the technical complexities in producing functionally mature DCs. The in vitro generation process, influenced by factors like the source of DCs, culture conditions, and maturation stimuli, is intricate and can significantly affect the therapy's success. Hence, the lack of standardized protocols for antigen selection and DC maturation further complicates the development of an effective therapy[284].

The prostate cancer TME presents another hurdle, often characterized by immunosuppressive elements that can impede the activity of cytotoxic T cells, undermining the effectiveness of DC therapy. Furthermore, even when initial immune responses are elicited, sustaining these responses over time remains a challenge, frequently leading to disease progression[285]. This lack of durable response necessitates repeated administrations or combination therapies, increasing treatment complexity and costs. The dynamic nature of the TME, with its evolving mechanisms of immunomodulation, makes it a moving target for DC therapy. Efforts to understand and manipulate this environment could be key to enhancing the therapy's effectiveness and durability.

DC therapy directly targeting the stromal compartment itself in prostate cancer offers a novel therapeutic avenue. For instance, immunotherapies directly targeting CAFs for depletion or reprogramming have the potential to reduce or eliminate tumor-promoting and immunosuppressive properties. However, this strategy faces challenges due to heterogeneity, phenotypic plasticity, and complex interactions within the tumor-stroma ecosystem, which can impede the efficacy of the immune response. Understanding and effectively manipulating these interactions are crucial for the success of DC therapy in targeting the stroma, potentially leading to more effective control of prostate cancer growth and metastases[286].

Despite being generally safe, DC therapy can induce adverse events like flu-like symptoms, injection site reactions, and potential autoimmunity[287,288]. These adverse events require careful monitoring to ensure patient safety. In conclusion, while DC therapy in prostate cancer represents a significant advancement in cancer immunotherapy, it is constrained by challenges such as inconsistent clinical responses, antigen selection, DC maturation difficulties, immune suppressive TME, transient immune responses, and safety issues. An important future approach should be focused on identifying biomarkers that could predict responses to DC therapy, thereby refining patient selection. Additionally, exploring synergies between DC therapy and other immunomodulatory approaches may unlock new avenues for more effective and comprehensive cancer treatment strategies.

SUMMARY

The survival of an organism is dependent on the maintenance of robust and dynamic systemic homeostatic mechanisms regulating physiological responses to both internal stimuli (wound repair, inflammation, and diseases) and external stimuli (food, pathogens, toxic pollutants, and drugs). Homeostasis is coordinated by the different functional systems within the body via a multitude of long-range (endocrine), short-range (paracrine, juxtacrine, neuronal signaling at synaptic junctions), and self (autocrine) cellular signaling. At the tissue level, the intracellular machinery of tissue-resident cells needs to coordinate and integrate complex signals from cellular and non-cellular components of the tissue environment. However, in the case of cancer, accumulating genomic or epigenetic aberrations in cancerous cells can decouple their functional interactions within a tissue, resulting in the development of neoplasia. Despite the dependence of cancer cells on multicellular interactions with the respective TME as well as systemic physiological environments, collectively referred to as "systems biology of cancer", conventional research continues to follow a reductionist approach, predominantly focusing on cancer-specific intracellular factors, overlooking broader systems influences on cancer pathobiology[279,280].

Although localized or locally advanced prostate cancer patients undergo definitive therapy with curative intent, up to 50% experience recurrence, progressing to mCRPC. While immunomodulatory therapies like ICIs have advanced as first- or second-line treatments, yielding promising results in various cancers, including non-small cell lung cancer (NSCLC) and colorectal cancer, their efficacy in advanced prostate cancer remains limited. Prostate cancer's immunologically "cold" TME underscores the need for a systems biology approach to identify and characterize spatial and temporal TME signatures. These signatures are crucial determinants in immunomodulation, disease progression, and treatment response[7,281-283].

In this review, we have highlighted the critical role of reactive stromal response in the evolution of cancer pathobiology through immunomodulation of the TME [Figure 2]. The reactive stromal response is an emergency/emergent response of the tissue to undergo rapid repair and reset homeostasis, a biological priority. The multifaceted role of reactive stromal cells includes their intercellular communication with tissue-resident and immune cells in the systemic circulation to enhance tissue repair while minimizing damage. Thus, to preserve homeostatic balance, the reactive stromal response encompasses immune-regulatory functions[284]. Therefore, adopting a cancer systems biology approach is crucial for fully grasping the dynamic interactions within the reactive/repair-centric TME. This methodology will enable researchers to identify key regulatory factors influencing cancer progression, survival, and resistance to immunotherapy, ultimately uncovering potential therapeutic targets. For instance, by integrating phenotypic and functional variations of the TME with molecular characteristics of cancer, an integrative model can be developed to delineate dynamic immunomodulation concurrent with tumor progression at the systems level. This approach will facilitate the discovery of new biomarkers that can be used for predicting immunotherapy response, aid in patient stratification, and inform on effective drug combinations to overcome drug resistance. However, a drawback of systems biology is its reliance on large sets of high-quality patient data collected over various time scales and concepts, which necessitates advanced downstream analyses and computations. Therefore, developing cost-effective and accessible technologies with user-friendly algorithms to integrate data from different omics platforms can revolutionize personalized cancer pathobiology modeling. These integrative, hypothesis-driven, and predictive models can advance our understanding of disease mechanisms and improve personalized treatment strategies[279,280].

DECLARATIONS

Authors’ Contributions

Conceptualized and designed the structural framework of the manuscript, performed comprehensive literature review, drafted the original manuscript, critically reviewed and revised the manuscript for intellectual content, approved the final version of the manuscript: Thomas R

Contributed to the conceptualization of the study; performed comprehensive literature review, and collaborated on writing of the manuscript: Jerome M, Krieger K

Contributed to the final revisions of the manuscript, critically reviewed the manuscript for intellectual content and accuracy of the information reported particularly in Tables 1 and 2: Jerome M, Krieger K, Ashraf N

Contributed significantly to the conceptualization of the manuscript, critically reviewed the content and provided crucial feedback on the manuscript, contributed to the comprehensive revisions of the initial and final draft of the manuscript, approved the final version of the manuscript: Rowley D

Availability of data and materials

Not applicable.

Financial support and sponsorship

This study was supported by National Cancer Institute of Health (NCI) (grant No: R01 CA221946, P30 CA125123), Early Investigator Research Award from the Department of Defense Prostate Cancer Research Program (W81WXH-21-1-0154), National Institutes of Health (K99MD018671), and CPRIT RP210027-Baylor College of Medicine Comprehensive Cancer Training Program.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

The Author(s) 2024.

REFERENCES

1. Cunha GR, Donjacour AA, Cooke PS, et al. The endocrinology and developmental biology of the prostate. Endocr Rev 1987;8:338-62.

2. Kumar VL, Majumder PK. Prostate gland: structure, functions and regulation. Int Urol Nephrol 1995;27:231-43.

3. Singh O, Bolla SR. Anatomy, abdomen and pelvis, prostate. Treasure Island: StatPearls; 2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK540987/ [Last accessed on 22 Jul 2024].

4. Rawla P. Epidemiology of prostate cancer. World J Oncol 2019;10:63-89.

5. Schrecengost R, Knudsen KE. Molecular pathogenesis and progression of prostate cancer. Semin Oncol 2013;40:244-58.

6. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12-49.

7. Shore ND, Moul JW, Pienta KJ, Czernin J, King MT, Freedland SJ. Biochemical recurrence in patients with prostate cancer after primary definitive therapy: treatment based on risk stratification. Prostate Cancer Prostatic Dis 2024;27:192-201.

8. Krušlin B, Ulamec M, Tomas D. Prostate cancer stroma: an important factor in cancer growth and progression. Bosn J Basic Med Sci 2015;15:1-8.

9. Levesque C, Nelson PS. Cellular constituents of the prostate stroma: key contributors to prostate cancer progression and therapy resistance. CSH Perspect Med 2018;8:a030510.

10. 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.

11. Barron DA, Rowley DR. The reactive stroma microenvironment and prostate cancer progression. Endocr Relat Cancer 2012;19:R187-204.

12. Davey RA, Grossmann M. Androgen receptor structure, function and biology: from bench to bedside. Clin Biochem Rev 2016;37:3-15.

13. Wen S, Chang HC, Tian J, Shang Z, Niu Y, Chang C. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. Am J Pathol 2015;185:293-301.

14. Cunha GR. Epithelio-mesenchymal interactions in primordial gland structures which become responsive to androgenic stimulation. Anat Rec 1972;172:179-95.

15. Schauer IG, Rowley DR. The functional role of reactive stroma in benign prostatic hyperplasia. Differentiation 2011;82:200-10.

16. Schauer IG, Ressler SJ, Tuxhorn JA, Dang TD, Rowley DR. Elevated epithelial expression of interleukin-8 correlates with myofibroblast reactive stroma in benign prostatic hyperplasia. Urology 2008;72:205-13.

17. Tuxhorn JA, Ayala GE, Smith MJ, Smith VC, Dang TD, Rowley DR. Reactive stroma in human prostate cancer: induction of myofibroblast phenotype and extracellular matrix remodeling. Clin Cancer Res 2002;8:2912-23.

18. Tuxhorn JA, Ayala GE, Rowley DR. Reactive stroma in prostate cancer progression. J Urol 2001;166:2472-83.

19. Kai F, Drain AP, Weaver VM. The extracellular matrix modulates the metastatic journey. Dev Cell 2019;49:332-46.

20. Valkenburg KC, de Groot AE, Pienta KJ. Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol 2018;15:366-81.

21. Puré E, Lo A. Can targeting stroma pave the way to enhanced antitumor immunity and immunotherapy of solid tumors? Cancer Immunol Res 2016;4:269-78.

22. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med 2013;19:1423-37.

23. Xiao Z, Todd L, Huang L, et al. Desmoplastic stroma restricts T cell extravasation and mediates immune exclusion and immunosuppression in solid tumors. Nat Commun 2023;14:5110.

24. Zhang FF, Qiao Y, Xie Y, et al. Epitope-based minigene vaccine targeting fibroblast activation protein α induces specific immune responses and anti-tumor effects in 4 T1 murine breast cancer model. Int Immunopharmacol 2022;112:109237.

25. Gorbach IN, Novikov DK. [Detection of leukocyte sensitization to tuberculosis mycobacterial antigens in newborns by the method of migration suppression in vitro]. Vopr Okhr Materin Det 1976;21:66-8.

26. Giacomini A, Grillo E, Rezzola S, et al. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev 2021;101:569-610.

27. Cunha GR. Mesenchymal-epithelial interactions: past, present, and future. Differentiation 2008;76:578-86.

28. Roberson KM, Edwards DW, Chang GC, Robertson CN. Isolation and characterization of a novel human prostatic stromal cell culture: DuK50. In Vitro Cell Dev Biol Anim 1995;31:840-5.

29. Webber MM, Trakul N, Thraves PS, et al. A human prostatic stromal myofibroblast cell line WPMY-1: a model for stromal-epithelial interactions in prostatic neoplasia. Carcinogenesis 1999;20:1185-92.

30. Simons BW, Hurley PJ, Huang Z, et al. Wnt signaling though beta-catenin is required for prostate lineage specification. Dev Biol 2012;371:246-55.

31. Farnsworth WE. Prostate stroma: physiology. Prostate 1999;38:60-72.

32. Mostofi FK, Sesterhenn IA, Davis CJ. A pathologist's view of prostatic carcinoma. Cancer 1993;71:906-32.

33. Steukers L, Glorieux S, Vandekerckhove AP, Favoreel HW, Nauwynck HJ. Diverse microbial interactions with the basement membrane barrier. Trends Microbiol 2012;20:147-55.

34. Brekken RA, Stupack D. Extracellular matrix in tumor biology. In: Biology of extracellular matrix. Cham: Springer International Publishing; 2017.

35. Welén K, Damber JE. Androgens, aging, and prostate health. Rev Endocr Metab Disord 2022;23:1221-31.

36. Stanworth RD, Jones TH. Testosterone for the aging male; current evidence and recommended practice. Clin Interv Aging 2008;3:25-44.

37. Prins GS, Huang L, Birch L, Pu Y. The role of estrogens in normal and abnormal development of the prostate gland. Ann N Y Acad Sci 2006;1089:1-13.

38. Shapiro E, Huang H, Masch RJ, McFadden DE, Wilson EL, Wu XR. Immunolocalization of estrogen receptor alpha and beta in human fetal prostate. J Urol 2005;174:2051-3.

39. Prins GS, Korach KS. The role of estrogens and estrogen receptors in normal prostate growth and disease. Steroids 2008;73:233-44.

40. Coffey DS, Walsh PC. Clinical and experimental studies of benign prostatic hyperplasia. Urol Clin North Am 1990;17:461-75.

41. Vickman RE, Franco OE, Moline DC, Vander Griend DJ, Thumbikat P, Hayward SW. The role of the androgen receptor in prostate development and benign prostatic hyperplasia: a review. Asian J Urol 2020;7:191-202.

42. Schulze H, Claus S. Histological localization of estrogen receptors in normal and diseased human prostates by immunocytochemistry. Prostate 1990;16:331-43.

43. Schulze H, Barrack ER. Immunocytochemical localization of estrogen receptors in the normal male and female canine urinary tract and prostate. Endocrinology 1987;121:1773-83.

44. Prins GS, Birch L. Neonatal estrogen exposure up-regulates estrogen receptor expression in the developing and adult rat prostate lobes. Endocrinology 1997;138:1801-9.

45. Yu Y, Liu L, Xie N, et al. Expression and function of the progesterone receptor in human prostate stroma provide novel insights to cell proliferation control. J Clin Endocrinol Metab 2013;98:2887-96.

46. Arora VK, Schenkein E, Murali R, et al. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell 2013;155:1309-22.

47. Tuong ZK, Loudon KW, Berry B, et al. Resolving the immune landscape of human prostate at a single-cell level in health and cancer. Cell Rep 2021;37:110132.

48. Henry GH, Malewska A, Joseph DB, et al. A cellular anatomy of the normal adult human prostate and prostatic urethra. Cell Rep 2018;25:3530-3542.e5.

49. Mass E, Ballesteros I, Farlik M, et al. Specification of tissue-resident macrophages during organogenesis. Science 2016;353:aaf4238.

50. Costello LC, Franklin RB. A comprehensive review of the role of zinc in normal prostate function and metabolism; and its implications in prostate cancer. Arch Biochem Biophys 2016;611:100-12.

51. Singh KK, Desouki MM, Franklin RB, Costello LC. Mitochondrial aconitase and citrate metabolism in malignant and nonmalignant human prostate tissues. Mol Cancer 2006;5:14.

52. Ginhoux F, Guilliams M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 2016;44:439-49.

53. Cannarella R, Condorelli RA, Barbagallo F, La Vignera S, Calogero AE. Endocrinology of the aging prostate: current concepts. Front Endocrinol 2021;12:554078.

54. Feldman HA, Longcope C, Derby CA, et al. Age trends in the level of serum testosterone and other hormones in middle-aged men: longitudinal results from the Massachusetts male aging study. J Clin Endocrinol Metab 2002;87:589-98.

55. O'Donnell AB, Araujo AB, McKinlay JB. The health of normally aging men: The massachusetts male aging study (1987-2004). Exp Gerontol 2004;39:975-84.

56. Huhtaniemi I. Late-onset hypogonadism: current concepts and controversies of pathogenesis, diagnosis and treatment. Asian J Androl 2014;16:192-202.

57. Rastrelli G, Vignozzi L, Corona G, Maggi M. Testosterone and benign prostatic hyperplasia. Sex Med Rev 2019;7:259-71.

58. Vignozzi L, Cellai I, Santi R, et al. Antiinflammatory effect of androgen receptor activation in human benign prostatic hyperplasia cells. J Endocrinol 2012;214:31-43.

59. Ho CH, Fan CK, Yu HJ, et al. Testosterone suppresses uropathogenic escherichia coli invasion and colonization within prostate cells and inhibits inflammatory responses through JAK/STAT-1 signaling pathway. PLoS One 2017;12:e0180244.

60. Quintar AA, Gonçalves BF, Taboga SR, Maldonado CA. The mongolian gerbil (meriones unguiculatus) as a model for inflammation-promoted prostate carcinogenesis. Cell Biol Int 2017;41:1234-8.

61. Cohen PG. Obesity in men: the hypogonadal-estrogen receptor relationship and its effect on glucose homeostasis. Med Hypotheses 2008;70:358-60.

62. Yang Y, Sheng J, Hu S, et al. Estrogen and G protein-coupled estrogen receptor accelerate the progression of benign prostatic hyperplasia by inducing prostatic fibrosis. Cell Death Dis 2022;13:533.

63. Ellem SJ, Risbridger GP. Aromatase and regulating the estrogen: androgen ratio in the prostate gland. J Steroid Biochem Mol Biol 2010;118:246-51.

64. Rebbeck TR. Prostate cancer disparities by race and ethnicity: from nucleotide to neighborhood. Cold Spring Harb Perspect Med 2018;8:a030387.

65. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin 2021;71:7-33.

66. Chowdhury-Paulino IM, Ericsson C, Vince R Jr, Spratt DE, George DJ, Mucci LA. Racial disparities in prostate cancer among black men: epidemiology and outcomes. Prostate Cancer Prostatic Dis 2022;25:397-402.

67. Hinata N, Fujisawa M. Racial differences in prostate cancer characteristics and cancer-specific mortality: an overview. World J Mens Health 2022;40:217-27.

68. Rohrmann S, Nelson WG, Rifai N, et al. Serum estrogen, but not testosterone, levels differ between black and white men in a nationally representative sample of Americans. J Clin Endocrinol Metab 2007;92:2519-25.

69. Henderson BE, Bernstein L, Ross RK, Depue RH, Judd HL. The early in utero oestrogen and testosterone environment of blacks and whites: potential effects on male offspring. Br J Cancer 1988;57:216-8.

70. Platz EA, Giovannucci E. The epidemiology of sex steroid hormones and their signaling and metabolic pathways in the etiology of prostate cancer. J Steroid Biochem Mol Biol 2004;92:237-53.

71. Giusti RM, Iwamoto K, Hatch EE. Diethylstilbestrol revisited: a review of the long-term health effects. Ann Intern Med 1995;122:778-88.

72. Fox JJ, Hashimoto T, Navarro HI, Garcia AJ, Shou BL, Goldstein AS. Highly multiplexed immune profiling throughout adulthood reveals kinetics of lymphocyte infiltration in the aging mouse prostate. Aging 2023;15:3356-80.

73. Khan D, Ansar Ahmed S. The immune system is a natural target for estrogen action: opposing effects of estrogen in two prototypical autoimmune diseases. Front Immunol 2015;6:635.

74. Eifert C, Powers RS. From cancer genomes to oncogenic drivers, tumour dependencies and therapeutic targets. Nat Rev Cancer 2012;12:572-8.

75. Dotto GP. Multifocal epithelial tumors and field cancerization: stroma as a primary determinant. J Clin Invest 2014;124:1446-53.

76. Cunha GR, Hayward SW, Wang YZ, Ricke WA. Role of the stromal microenvironment in carcinogenesis of the prostate. Int J Cancer 2003;107:1-10.

77. Cunha GR, Hayward SW, Wang YZ. Role of stroma in carcinogenesis of the prostate. Differentiation 2002;70:473-85.

78. Wang Y, Sudilovsky D, Zhang B, et al. A human prostatic epithelial model of hormonal carcinogenesis. Cancer Res 2001;61:6064-72.

79. Ricke WA, Ishii K, Ricke EA, et al. Steroid hormones stimulate human prostate cancer progression and metastasis. Int J Cancer 2006;118:2123-31.

80. Hayward SW, Wang Y, Cao M, et al. Malignant transformation in a nontumorigenic human prostatic epithelial cell line. Cancer Res 2001;61:8135-42.

81. Hu B, Castillo E, Harewood L, et al. Multifocal epithelial tumors and field cancerization from loss of mesenchymal CSL signaling. Cell 2012;149:1207-20.

82. Salem AF, Al-Zoubi MS, Whitaker-Menezes D, et al. Cigarette smoke metabolically promotes cancer, via autophagy and premature aging in the host stromal microenvironment. Cell Cycle 2013;12:818-25.

83. Yoshimoto S, Loo TM, Atarashi K, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013;499:97-101.

84. Berglund E, Maaskola J, Schultz N, et al. Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of heterogeneity. Nat Commun 2018;9:2419.

85. Kwon OJ, Zhang Y, Li Y, et al. Functional heterogeneity of mouse prostate stromal cells revealed by single-cell RNA-Seq. iScience 2019;13:328-38.

86. Huet E, Jaroz C, Nguyen HQ, et al. Stroma in normal and cancer wound healing. FEBS J 2019;286:2909-20.

87. Varga J, Brenner DA, Phan SH. Fibrosis research: methods and protocols. Berlin: Springer; 2008.

88. Clark RAF. The molecular and cellular biology of wound repair. New York: Plenum Press;1996

89. Schäfer M, Werner S. Cancer as an overhealing wound: an old hypothesis revisited. Nat Rev Mol Cell Biol 2008;9:628-38.

90. Haddow A. Molecular repair, wound healing, and carcinogenesis: tumor production a possible overhealing? Amsterdam: Elsevier; 1973. pp. 181-234.

91. Langley RR, Fidler IJ. The seed and soil hypothesis revisited--the role of tumor-stroma interactions in metastasis to different organs. Int J Cancer 2011;128:2527-35.

92. Plava J, Cihova M, Burikova M, Matuskova M, Kucerova L, Miklikova S. Recent advances in understanding tumor stroma-mediated chemoresistance in breast cancer. Mol Cancer 2019;18:67.

93. Mesker WE, Junggeburt JM, Szuhai K, et al. The carcinoma-stromal ratio of colon carcinoma is an independent factor for survival compared to lymph node status and tumor stage. Cell Oncol 2007;29:387-98.

94. Almangush A, Alabi RO, Troiano G, et al. Clinical significance of tumor-stroma ratio in head and neck cancer: a systematic review and meta-analysis. BMC Cancer 2021;21:480.

95. He R, Li D, Liu B, et al. The prognostic value of tumor-stromal ratio combined with TNM staging system in esophagus squamous cell carcinoma. J Cancer 2021;12:1105-14.

96. Zhu Y, Jin Z, Qian Y, Shen Y, Wang Z. Prognostic value of tumor-stroma ratio in rectal cancer: a systematic review and meta-analysis. Front Oncol 2021;11:685570.

97. Ruder S, Gao Y, Ding Y, et al. Development and validation of a quantitative reactive stroma biomarker (qRS) for prostate cancer prognosis. Hum Pathol 2022;122:84-91.

98. Ayala G, Tuxhorn JA, Wheeler TM, et al. Reactive stroma as a predictor of biochemical-free recurrence in prostate cancer. Clin Cancer Res 2003;9:4792-801.

99. Tran LL, Dang T, Thomas R, Rowley DR. ELF3 mediates IL-1α induced differentiation of mesenchymal stem cells to inflammatory iCAFs. Stem Cells 2021;39:1766-77.

100. Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res 1999;59:5002-11.

101. Hayashi N, Cunha GR. Mesenchyme-induced changes in the neoplastic characteristics of the Dunning prostatic adenocarcinoma. Cancer Res 1991;51:4924-30.

102. Proia DA, Kuperwasser C. Stroma: tumor agonist or antagonist. Cell Cycle 2005;4:1022-5.

103. Mun JY, Leem SH, Lee JH, Kim HS. Dual relationship between stromal cells and immune cells in the tumor microenvironment. Front Immunol 2022;13:864739.

104. Sanjabi S, Oh SA, Li MO. Regulation of the immune response by TGF-β: from conception to autoimmunity and infection. Cold Spring Harb Perspect Biol 2017;9:a022236.

105. Zhang F, Wang H, Wang X, et al. TGF-β induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget 2016;7:52294-306.

106. Penn JW, Grobbelaar AO, Rolfe KJ. The role of the TGF-beta family in wound healing, burns and scarring: a review. Int J Burns Trauma 2012;2:18-28.

107. Zhang Y, Alexander PB, Wang XF. TGF-β family signaling in the control of cell proliferation and survival. Cold Spring Harb Perspect Biol 2017;9:a022145.

108. Xu J, Lamouille S, Derynck R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res 2009;19:156-72.

109. Farooq M, Khan AW, Kim MS, Choi S. The role of fibroblast growth factor (FGF) signaling in tissue repair and regeneration. Cells 2021;10:3242.

110. Pierce GF, Mustoe TA, Altrock BW, Deuel TF, Thomason A. Role of platelet-derived growth factor in wound healing. J Cell Biochem 1991;45:319-26.

111. Diller RB, Tabor AJ. The role of the extracellular matrix (ECM) in wound healing: a review. Biomimetics 2022;7:87.

112. Rodriguez-Pascual F, Rosell-Garcia T. Lysyl oxidases: functions and disorders. J Glaucoma 2018;27 Suppl 1:S15-9.

113. Wang LC, Lo A, Scholler J, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res 2014;2:154-66.

114. Nissen NI, Karsdal M, Willumsen N. Collagens and cancer associated fibroblasts in the reactive stroma and its relation to cancer biology. J Exp Clin Cancer Res 2019;38:115.

115. Henke E, Nandigama R, Ergün S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci 2019;6:160.

116. Hughes CC. Endothelial-stromal interactions in angiogenesis. Curr Opin Hematol 2008;15:204-9.

117. Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag 2006;2:213-9.

118. Li Y, Zhao L, Li XF. Hypoxia and the tumor microenvironment. Technol Cancer Res Treat 2021;20:15330338211036304.

119. Zhang Y, Coleman M, Brekken RA. Perspectives on hypoxia signaling in tumor stroma. Cancers 2021;13:3070.

120. Crola Da Silva C, Lamerant-Fayel N, Paprocka M, et al. Selective human endothelial cell activation by chemokines as a guide to cell homing. Immunology 2009;126:394-404.

121. Crowley T, Buckley CD, Clark AR. Stroma: the forgotten cells of innate immune memory. Clin Exp Immunol 2018;193:24-36.

122. Jiang Z, Zhou J, Li L, et al. Pericytes in the tumor microenvironment. Cancer Lett 2023;556:216074.

123. Thomas H, Cowin AJ, Mills SJ. The importance of pericytes in healing: wounds and other pathologies. Int J Mol Sci 2017;18:1129.

124. Paiva AE, Lousado L, Guerra DAP, et al. Pericytes in the premetastatic niche. Cancer Res 2018;78:2779-86.

125. Kirk T, Ahmed A, Rognoni E. Fibroblast memory in development, homeostasis and disease. Cells 2021;10:2840.

126. Netea MG, Quintin J, van der Meer JW. Trained immunity: a memory for innate host defense. Cell Host Microbe 2011;9:355-61.

127. Wang T, Hu Y, Dusi S, et al. "Open Sesame" to the complexity of pattern recognition receptors of myeloid-derived suppressor cells in cancer. Front Immunol 2023;14:1130060.

128. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 2006;124:783-801.

129. Netea MG, van der Meer JW. Trained immunity: an ancient way of remembering. Cell Host Microbe 2017;21:297-300.

130. Dakin SG, Buckley CD, Al-Mossawi MH, et al. Persistent stromal fibroblast activation is present in chronic tendinopathy. Arthritis Res Ther 2017;19:16.

131. Klein K, Frank-Bertoncelj M, Karouzakis E, et al. The epigenetic architecture at gene promoters determines cell type-specific LPS tolerance. J Autoimmun 2017;83:122-33.

132. Sohn C, Lee A, Qiao Y, Loupasakis K, Ivashkiv LB, Kalliolias GD. Prolonged tumor necrosis factor α primes fibroblast-like synoviocytes in a gene-specific manner by altering chromatin. Arthritis Rheumatol 2015;67:86-95.

133. Koch SR, Lamb FS, Hellman J, Sherwood ER, Stark RJ. Potentiation and tolerance of toll-like receptor priming in human endothelial cells. Transl Res 2017;180:53-67.e4.

134. Naik S, Larsen SB, Gomez NC, et al. Author correction: inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 2018;560:E2.

135. Ara T, Kurata K, Hirai K, et al. Human gingival fibroblasts are critical in sustaining inflammation in periodontal disease. J Periodontal Res 2009;44:21-7.

136. Lee A, Qiao Y, Grigoriev G, et al. Tumor necrosis factor α induces sustained signaling and a prolonged and unremitting inflammatory response in rheumatoid arthritis synovial fibroblasts. Arthritis Rheum 2013;65:928-38.

137. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 1994;76:301-14.

138. Wolff B, Burns AR, Middleton J, Rot A. Endothelial cell "memory" of inflammatory stimulation: human venular endothelial cells store interleukin 8 in Weibel-Palade bodies. J Exp Med 1998;188:1757-62.

139. Bonfanti R, Furie BC, Furie B, Wagner DD. PADGEM (GMP140) is a component of Weibel-Palade bodies of human endothelial cells. Blood 1989;73:1109-12.

140. McEver RP, Beckstead JH, Moore KL, Marshall-Carlson L, Bainton DF. GMP-140, a platelet alpha-granule membrane protein, is also synthesized by vascular endothelial cells and is localized in Weibel-Palade bodies. J Clin Invest 1989;84:92-9.

141. Zimmerman GA, McIntyre TM, Mehra M, Prescott SM. Endothelial cell-associated platelet-activating factor: a novel mechanism for signaling intercellular adhesion. J Cell Biol 1990;110:529-40.

142. Séguin C, Abid MR, Spokes KC, et al. Priming effect of homocysteine on inducible vascular cell adhesion molecule-1 expression in endothelial cells. Biomed Pharmacother 2008;62:395-400.

143. Ceriello A, Ihnat MA, Thorpe JE. Clinical review 2: the "metabolic memory": is more than just tight glucose control necessary to prevent diabetic complications? J Clin Endocrinol Metab 2009;94:410-5.

144. Testa R, Bonfigli AR, Prattichizzo F, La Sala L, De Nigris V, Ceriello A. The "Metabolic Memory" theory and the early treatment of hyperglycemia in prevention of diabetic complications. Nutrients 2017;9:437.

145. Yao Y, Song Q, Hu C, et al. Endothelial cell metabolic memory causes cardiovascular dysfunction in diabetes. Cardiovasc Res 2022;118:196-211.

146. Muhl L, Genové G, Leptidis S, et al. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat Commun 2020;11:3953.

147. Amersfoort J, Eelen G, Carmeliet P. Immunomodulation by endothelial cells - partnering up with the immune system? Nat Rev Immunol 2022;22:576-88.

148. Yeo SY, Lee KW, Shin D, An S, Cho KH, Kim SH. A positive feedback loop bi-stably activates fibroblasts. Nat Commun 2018;9:3016.

149. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 2008;214:199-210.

150. Rosenberg SA. Raising the bar: the curative potential of human cancer immunotherapy. Sci Transl Med 2012;4:127ps8.

151. Lin R, Zhang C, Zheng J, et al. Chronic inflammation-associated genomic instability paves the way for human esophageal carcinogenesis. Oncotarget 2016;7:24564-71.

152. Hibino S, Kawazoe T, Kasahara H, et al. Inflammation-induced tumorigenesis and metastasis. Int J Mol Sci 2021;22:5421.

153. Bockerstett KA, DiPaolo RJ. Regulation of gastric carcinogenesis by inflammatory cytokines. Cell Mol Gastroenter Hepatol 2017;4:47-53.

154. Qian S, Golubnitschaja O, Zhan X. Chronic inflammation: key player and biomarker-set to predict and prevent cancer development and progression based on individualized patient profiles. EPMA J 2019;10:365-81.

155. He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B 2021;11:3379-92.

156. Ashrafi A, Akter Z, Modareszadeh P, et al. Current landscape of therapeutic resistance in lung cancer and promising strategies to overcome resistance. Cancers 2022;14:4562.

157. Marin JJ, Al-Abdulla R, Lozano E, et al. Mechanisms of resistance to chemotherapy in gastric cancer. Anti Agent Med Chem 2016;16:318-34.

158. Wang Q, Shen X, Chen G, Du J. Drug resistance in colorectal cancer: from mechanism to clinic. Cancers 2022;14:2928.

159. Scott AK, Rafuse M, Neu CP. Mechanically induced alterations in chromatin architecture guide the balance between cell plasticity and mechanical memory. Front Cell Dev Biol 2023;11:1084759.

160. Burnet FM. The Concept of immunological surveillance. Prog Exp Tumor Res 1970;13:1-27.

161. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002;3:991-8.

162. Penn I. Posttransplant malignancies. Transplant Proc 1999;31:1260-2.

163. Sheil AG. Cancer after transplantation. World J Surg 1986;10:389-96.

164. Penn I. Malignant melanoma in organ allograft recipients. Transplantation 1996;61:274-8.

165. Penn I. Sarcomas in organ allograft recipients. Transplantation 1995;60:1485-91.

166. Pham SM, Kormos RL, Landreneau RJ, et al. Solid tumors after heart transplantation: lethality of lung cancer. Ann Thorac Surg 1995;60:1623-6.

167. Nair SS, Weil R, Dovey Z, Davis A, Tewari AK. The tumor microenvironment and immunotherapy in prostate and bladder cancer. Urol Clin North Am 2020;47:e17-54.

168. Zhong C, Li Y, Yang J, et al. Immunotherapy for hepatocellular carcinoma: current limits and prospects. Front Oncol 2021;11:589680.

169. Jia D, Zhou Z, Kwon OJ, et al. Stromal FOXF2 suppresses prostate cancer progression and metastasis by enhancing antitumor immunity. Nat Commun 2022;13:6828.

170. Lander VE, Belle JI, Kingston NL, et al. Stromal reprogramming by FAK inhibition overcomes radiation resistance to allow for immune priming and response to checkpoint blockade. Cancer Discov 2022;12:2774-99.

171. Yang D, Duan MH, Yuan QE, et al. Suppressive stroma-immune prognostic signature impedes immunotherapy in ovarian cancer and can be reversed by PDGFRB inhibitors. J Transl Med 2023;21:586.

172. Bremnes RM, Dønnem T, Al-Saad S, et al. The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol 2011;6:209-17.

173. Nishikawa H, Koyama S. Mechanisms of regulatory T cell infiltration in tumors: implications for innovative immune precision therapies. J Immunother Cancer 2021;9:e002591.

174. Lunardi S, Jamieson NB, Lim SY, et al. IP-10/CXCL10 induction in human pancreatic cancer stroma influences lymphocytes recruitment and correlates with poor survival. Oncotarget 2014;5:11064-80.

175. Vilgelm AE, Richmond A. Chemokines modulate immune surveillance in tumorigenesis, metastasis, and response to immunotherapy. Front Immunol 2019;10:333.

176. Hussain S, Peng B, Cherian M, Song JW, Ahirwar DK, Ganju RK. The roles of stroma-derived chemokine in different stages of cancer metastases. Front Immunol 2020;11:598532.

177. Deng J, Jiang R, Meng E, Wu H. CXCL5: a coachman to drive cancer progression. Front Oncol 2022;12:944494.

178. Kraman M, Bambrough PJ, Arnold JN, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010;330:827-30.

179. Poznansky MC, Olszak IT, Foxall R, Evans RH, Luster AD, Scadden DT. Active movement of T cells away from a chemokine. Nat Med 2000;6:543-8.

180. Kryczek I, Wei S, Keller E, Liu R, Zou W. Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am J Physiol Cell Physiol 2007;292:C987-95.

181. Schreiber H, Rowley DA. Cancer. Awakening immunity. Science 2010;330:761-2.

182. Yang L, Pang Y, Moses HL. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol 2010;31:220-7.

183. Shi X, Young CD, Zhou H, Wang X. Transforming growth factor-β signaling in fibrotic diseases and cancer-associated fibroblasts. Biomolecules 2020;10:1666.

184. Mariathasan S, Turley SJ, Nickles D, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544-8.

185. Zippi M, De Toma G, Minervini G, et al. Desmoplasia influenced recurrence of disease and mortality in stage III colorectal cancer within five years after surgery and adjuvant therapy. Saudi J Gastroenterol 2017;23:39-44.

186. Whatcott CJ, Posner RG, Von Hoff DD, Han H. Chapter 8 desmoplasia and chemoresistance in pancreatic cancer. In: Grippo PJ, Munshi HG, editors. Pancreatic cancer and tumor microenvironment. India: Trivandrum; 2012. Available from: https://www.ncbi.nlm.nih.gov/books/NBK98939/ [Last accessed on 22 Jul 2024].

187. DeClerck YA. Desmoplasia: a response or a niche? Cancer Discov 2012;2:772-4.

188. González-González L, Alonso J. Periostin: a matricellular protein with multiple functions in cancer development and progression. Front Oncol 2018;8:225.

189. Wei T, Wang K, Liu S, et al. Periostin deficiency reduces PD-1+ tumor-associated macrophage infiltration and enhances anti-PD-1 efficacy in colorectal cancer. Cell Rep 2023;42:112090.

190. Mortezaee K. Immune escape: a critical hallmark in solid tumors. Life Sci 2020;258:118110.

191. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197-218.

192. Hussein MR, Al-Assiri M, Musalam AO. Phenotypic characterization of the infiltrating immune cells in normal prostate, benign nodular prostatic hyperplasia and prostatic adenocarcinoma. Exp Mol Pathol 2009;86:108-13.

193. von Amsberg G, Alsdorf W, Karagiannis P, et al. Immunotherapy in advanced prostate cancer-light at the end of the tunnel? Int J Mol Sci 2022;23:2569.

194. Anker JF, Naseem AF, Mok H, Schaeffer AJ, Abdulkadir SA, Thumbikat P. Multi-faceted immunomodulatory and tissue-tropic clinical bacterial isolate potentiates prostate cancer immunotherapy. Nat Commun 2018;9:1591.

195. Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer 2018;6:157.

196. Wang I, Song L, Wang BY, Rezazadeh Kalebasty A, Uchio E, Zi X. Prostate cancer immunotherapy: a review of recent advancements with novel treatment methods and efficacy. Am J Clin Exp Urol 2022;10:210-33.

197. Ma Z, Zhang W, Dong B, et al. Docetaxel remodels prostate cancer immune microenvironment and enhances checkpoint inhibitor-based immunotherapy. Theranostics 2022;12:4965-79.

198. Chesner L, Graff J, Polesso F, et al. Abstract B041: AR suppresses MHC class I expression and T-cell response in prostate cancer. Cancer Res 2023;83:B041.

199. Kogan-Sakin I, Cohen M, Paland N, et al. Prostate stromal cells produce CXCL-1, CXCL-2, CXCL-3 and IL-8 in response to epithelia-secreted IL-1. Carcinogenesis 2009;30:698-705.

200. Tse BW, Scott KF, Russell PJ. Paradoxical roles of tumour necrosis factor-alpha in prostate cancer biology. Prostate Cancer 2012;2012:128965.

201. Smith BN, Mishra R, Billet S, et al. Antagonizing CD105 and androgen receptor to target stromal-epithelial interactions for clinical benefit. Mol Ther 2023;31:78-89.

202. Zhou C, Gao Y, Ding P, Wu T, Ji G. The role of CXCL family members in different diseases. Cell Death Discov 2023;9:212.

203. Bullock K, Richmond A. Suppressing MDSC recruitment to the tumor microenvironment by antagonizing CXCR2 to enhance the efficacy of immunotherapy. Cancers 2021;13:6293.

204. Korbecki J, Kupnicka P, Chlubek M, Gorący J, Gutowska I, Baranowska-Bosiacka I. CXCR2 receptor: regulation of expression, signal transduction, and involvement in cancer. Int J Mol Sci 2022;23:2168.

205. Di Mitri D, Mirenda M, Vasilevska J, et al. Re-education of tumor-associated macrophages by CXCR2 blockade drives senescence and tumor inhibition in advanced prostate cancer. Cell Rep 2019;28:2156-2168.e5.

206. Bahig H, Taussky D, Delouya G, et al. Neutrophil count is associated with survival in localized prostate cancer. BMC Cancer 2015;15:594.

207. Sharma J, Gray KP, Harshman LC, et al. Elevated IL-8, TNF-α, and MCP-1 in men with metastatic prostate cancer starting androgen-deprivation therapy (ADT) are associated with shorter time to castration-resistance and overall survival. Prostate 2014;74:820-8.

208. Minas TZ, Candia J, Dorsey TH, et al. Serum proteomics links suppression of tumor immunity to ancestry and lethal prostate cancer. Nat Commun 2022;13:1759.

209. Wallace TA, Prueitt RL, Yi M, et al. Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res 2008;68:927-36.

210. Zhu W, Wu J, Huang J, et al. Multi-omics analysis reveals a macrophage-related marker gene signature for prognostic prediction, immune landscape, genomic heterogeneity, and drug choices in prostate cancer. Front Immunol 2023;14:1122670.

211. Chen C, Luo J, Wang X. Identification of prostate cancer subtypes based on immune signature scores in bulk and single-cell transcriptomes. Med Oncol 2022;39:123.

212. Guo T, Wang J, Yan S, et al. A combined signature of glycolysis and immune landscape predicts prognosis and therapeutic response in prostate cancer. Front Endocrinol 2022;13:1037099.

213. Ren C, Wang Q, Wang S, et al. Metabolic syndrome-related prognostic index: predicting biochemical recurrence and differentiating between cold and hot tumors in prostate cancer. Front Endocrinol 2023;14:1148117.

214. Li N, Yu K, Lin Z, Zeng D. Development of a novel immune subtyping system expanded with immune landscape and an 11-gene signature for predicting prostate cancer survival. J Oncol 2022;2022:1183173.

215. Keam SP, Halse H, Nguyen T, et al. High dose-rate brachytherapy of localized prostate cancer converts tumors from cold to hot. J Immunother Cancer 2020;8:e000792.

216. Shen Y, Xu H, Long M, et al. Screening to identify an immune landscape-based prognostic predictor and therapeutic target for prostate cancer. Front Oncol 2021;11:761643.

217. Zhang G, Luo Y. An immune-related lncRNA signature to predict the biochemical recurrence and immune landscape in prostate cancer. Int J Gen Med 2021;14:9031-49.

218. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74.

219. Antognelli C, Mandarano M, Prosperi E, Sidoni A, Talesa VN. Glyoxalase-1-dependent methylglyoxal depletion sustains PD-L1 expression in metastatic prostate cancer cells: a novel mechanism in cancer immunosurveillance escape and a potential novel target to overcome PD-L1 blockade resistance. Cancers 2021;13:2965.

220. Datta M, Coussens LM, Nishikawa H, Hodi FS, Jain RK. Reprogramming the tumor microenvironment to improve immunotherapy: emerging strategies and combination therapies. Am Soc Clin Oncol Educ Book 2019;39:165-74.

221. Perera MPJ, Thomas PB, Risbridger GP, et al. Chimeric antigen receptor T-cell therapy in metastatic castrate-resistant prostate cancer. Cancers 2022;14:503.

222. Bander NH, Nanus DM, Milowsky MI, Kostakoglu L, Vallabahajosula S, Goldsmith SJ. Targeted systemic therapy of prostate cancer with a monoclonal antibody to prostate-specific membrane antigen. Semin Oncol 2003;30:667-76.

223. Sardinha M, Palma Dos Reis AF, Barreira JV, Fontes Sousa M, Pacey S, Luz R. Antibody-drug conjugates in prostate cancer: a systematic review. Cureus 2023;15:e34490.

224. Kamat NV, Yu EY, Lee JK. BiTE-ing into prostate cancer with bispecific T-cell engagers. Clin Cancer Res 2021;27:2675-7.

225. Yang K, Feng S, Luo Z. Oncolytic adenovirus, a new treatment strategy for prostate cancer. Biomedicines 2022;10:3262.

226. Pieczonka CM, Telonis D, Mouraviev V, Albala D. Sipuleucel-T for the treatment of patients with metastatic castrate-resistant prostate cancer: considerations for clinical practice. Rev Urol 2015;17:203-10.

227. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411-22.

228. Donninger H, Li C, Eaton JW, Yaddanapudi K. Cancer vaccines: promising therapeutics or an unattainable dream. Vaccines 2021;9:668.

229. Chen S, Wainwright DA, Wu JD, et al. CD73: an emerging checkpoint for cancer immunotherapy. Immunotherapy 2019;11:983-97.

230. Mbongue JC, Nicholas DA, Torrez TW, Kim NS, Firek AF, Langridge WH. The role of indoleamine 2, 3-dioxygenase in immune suppression and autoimmunity. Vaccines 2015;3:703-29.

231. Solinas C, Migliori E, De Silva P, Willard-Gallo K. LAG3: the biological processes that motivate targeting this immune checkpoint molecule in human cancer. Cancers 2019;11:1213.

232. Miller RA, Luke JJ, Hu S, et al. Anti-CD73 antibody activates human B cells, enhances humoral responses and induces redistribution of B cells in patients with cancer. J Immunother Cancer 2022;10:e005802.

233. Fan X, Quezada SA, Sepulveda MA, Sharma P, Allison JP. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med 2014;211:715-25.

234. Fu Y, Lin Q, Zhang Z, Zhang L. Therapeutic strategies for the costimulatory molecule OX40 in T-cell-mediated immunity. Acta Pharm Sin B 2020;10:414-33.

235. Ye L, Jia K, Wang L, et al. CD137, an attractive candidate for the immunotherapy of lung cancer. Cancer Sci 2020;111:1461-7.

236. Bayer AL, Pugliese A, Malek TR. The IL-2/IL-2R system: from basic science to therapeutic applications to enhance immune regulation. Immunol Res 2013;57:197-209.

237. Schepisi G, Cursano MC, Casadei C, et al. CAR-T cell therapy: a potential new strategy against prostate cancer. J Immunother Cancer 2019;7:258.

238. Patel S, Burga RA, Powell AB, et al. Beyond CAR T cells: other cell-based immunotherapeutic strategies against cancer. front oncol 2019;9:196.

239. Mills JK, Henderson MA, Giuffrida L, et al. Generating CAR T cells from tumor-infiltrating lymphocytes. Ther Adv Vaccines Immunother 2021;9:25151355211017119.

240. Zhou B, Lin W, Long Y, et al. Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022;7:95.

241. Day KC, Lorenzatti Hiles G, Kozminsky M, et al. HER2 and EGFR overexpression support metastatic progression of prostate cancer to bone. Cancer Res 2017;77:74-85.

242. Hsu EC, Rice MA, Bermudez A, et al. Trop2 is a driver of metastatic prostate cancer with neuroendocrine phenotype via PARP1. Proc Natl Acad Sci USA 2020;117:2032-42.

243. Rosellini M, Santoni M, Mollica V, et al. Treating prostate cancer by antibody-drug conjugates. Int J Mol Sci 2021;22:1551.

244. Tian Z, Liu M, Zhang Y, Wang X. Bispecific T cell engagers: an emerging therapy for management of hematologic malignancies. J Hematol Oncol 2021;14:75.

245. Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023;8:156.

246. Lee J, Fassnacht M, Nair S, Boczkowski D, Gilboa E. Tumor immunotherapy targeting fibroblast activation protein, a product expressed in tumor-associated fibroblasts. Cancer Res 2005;65:11156-63.

247. Wen Y, Wang CT, Ma TT, et al. Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci 2010;101:2325-32.

248. Yu F, Wang X, Guo ZS, Bartlett DL, Gottschalk SM, Song XT. T-cell engager-armed oncolytic vaccinia virus significantly enhances antitumor therapy. Mol Ther 2014;22:102-11.

249. Freedman JD, Duffy MR, Lei-Rossmann J, et al. An oncolytic virus expressing a T-cell engager simultaneously targets cancer and immunosuppressive stromal cells. Cancer Res 2018;78:6852-65.

250. Chaudary N, Pintilie M, Jelveh S, Lindsay P, Hill RP, Milosevic M. Plerixafor improves primary tumor response and reduces metastases in cervical cancer treated with radio-chemotherapy. Clin Cancer Res 2017;23:1242-9.

251. Kim ST, Hong JY, Park SH, et al. First-in-human phase I trial of anti-hepatocyte growth factor antibody (YYB101) in refractory solid tumor patients. Ther Adv Med Oncol 2020;12:1758835920926796.

252. Zhang X, Luo H. Effects of thalidomide on growth and VEGF-A expression in SW480 colon cancer cells. Oncol Lett 2018;15:3313-20.

253. Pulte ED, Dmytrijuk A, Nie L, et al. FDA approval summary: lenalidomide as maintenance therapy after autologous stem cell transplant in newly diagnosed multiple myeloma. Oncologist 2018;23:734-9.

254. Summers J, Cohen MH, Keegan P, Pazdur R. FDA drug approval summary: bevacizumab plus interferon for advanced renal cell carcinoma. Oncologist 2010;15:104-11.

255. Singh AD, Parmar S. Ramucirumab (Cyramza): a breakthrough treatment for gastric cancer. P T 2015;40:430-68.

256. Chiorean EG, Sweeney C, Youssoufian H, et al. A phase I study of olaratumab, an anti-platelet-derived growth factor receptor alpha (PDGFRα) monoclonal antibody, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2014;73:595-604.

257. Kwon MJ. Matrix metalloproteinases as therapeutic targets in breast cancer. Front Oncol 2022;12:1108695.

258. Nadal L, Corbellari R, Villa A, et al. Novel human monoclonal antibodies specific to the alternatively spliced domain D of Tenascin C efficiently target tumors in vivo. MAbs 2020;12:1836713.

259. Neesse A, Frese KK, Bapiro TE, et al. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proc Natl Acad Sci USA 2013;110:12325-30.

260. Ke FY, Chen WY, Lin MC, Hwang YC, Kuo KT, Wu HC. Novel monoclonal antibody against integrin α3 shows therapeutic potential for ovarian cancer. Cancer Sci 2020;111:3478-92.

261. Sun Y, Zhao C, Ye Y, et al. High expression of fibronectin 1 indicates poor prognosis in gastric cancer. Oncol Lett 2020;19:93-102.

262. Streel G, Lucas S. Targeting immunosuppression by TGF-β1 for cancer immunotherapy. Biochem Pharmacol 2021;192:114697.

263. Ma C, Xi S, Sun H, Zhang M, Pei Y. Identifying the oncogenic roles of FAP in human cancers based on systematic analysis. Aging 2023;15:7056-83.

264. Martori C, Sanchez-Moral L, Paul T, et al. Macrophages as a therapeutic target in metastatic prostate cancer: a way to overcome immunotherapy resistance? Cancers 2022;14:440.

265. Gordon S, Plüddemann A. The mononuclear phagocytic system. generation of diversity. Front Immunol 2019;10:1893.

266. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 2014;6:13.

267. Das A, Sinha M, Datta S, et al. Monocyte and macrophage plasticity in tissue repair and regeneration. Am J Pathol 2015;185:2596-606.

268. Gordon S, Plüddemann A. Tissue macrophages: heterogeneity and functions. BMC Biol 2017;15:53.

269. He MX, Cuoco MS, Crowdis J, et al. Transcriptional mediators of treatment resistance in lethal prostate cancer. Nat Med 2021;27:426-33.

270. Stout RD, Jiang C, Matta B, Tietzel I, Watkins SK, Suttles J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J Immunol 2005;175:342-9.

271. Li XF, Selli C, Zhou HL, et al. Macrophages promote anti-androgen resistance in prostate cancer bone disease. J Exp Med 2023:220.

272. Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov 2022;21:799-820.

273. Abram CL, Lowell CA. The diverse functions of Src family kinases in macrophages. Front Biosci 2008;13:4426-50.

274. Loi M, Salvatore G, Sottili M, et al. Tumor-associated macrophages (TAMs) modulate response to HER2-targeted agents in a humanized mouse model of breast cancer. Clin Transl Oncol 2022;24:1395-402.

275. Watanabe H, Ohashi K, Nishii K, et al. A long-term response to nivolumab in a case of PD-L1-negative lung adenocarcinoma with an EGFR mutation and surrounding PD-L1-positive tumor-associated macrophages. Intern Med 2019;58:3033-7.

276. Su S, Lei A, Wang X, et al. Induced CAR-macrophages as a novel therapeutic cell type for cancer immune cell therapies. Cells 2022;11:1652.

277. Liu M, Liu J, Liang Z, et al. CAR-macrophages and CAR-T cells synergistically kill tumor cells in vitro. Cells 2022;11:3692.

278. Huber ML, Haynes L, Parker C, Iversen P. Interdisciplinary critique of sipuleucel-T as immunotherapy in castration-resistant prostate cancer. J Natl Cancer Inst 2012;104:273-9.

279. Murphy G, Tjoa B, Ragde H, Kenny G, Boynton A. Phase I clinical trial: T-cell therapy for prostate cancer using autologous dendritic cells pulsed with HLA-A0201-specific peptides from prostate-specific membrane antigen. Prostate 1996;29:371-80.

280. Tjoa B, Erickson S, Bowes V, et al. Follow-up evaluation of prostate cancer patients infused with autologous dendritic cells pulsed with PSMA peptides. Prostate 1997;32:272-8.

281. Jähnisch H, Füssel S, Kiessling A, et al. Dendritic cell-based immunotherapy for prostate cancer. Clin Dev Immunol 2010;2010:517493.

282. Kiessling A, Wehner R, Füssel S, Bachmann M, Wirth MP, Schmitz M. Tumor-associated antigens for specific immunotherapy of prostate cancer. Cancers 2012;4:193-217.

283. Kitamura H, Torigoe T, Asanuma H, Honma I, Sato N, Tsukamoto T. Down-regulation of HLA class I antigens in prostate cancer tissues and up-regulation by histone deacetylase inhibition. J Urol 2007;178:692-6.

284. Lozano M, Cid J, Benitez-Ribas D, Otero MJ. Technical challenges in the manufacture of dendritic cell cancer therapies. Eur Oncol Haematol 2019;15:22-8.

285. Kongsted P, Borch TH, Ellebaek E, et al. Dendritic cell vaccination in combination with docetaxel for patients with metastatic castration-resistant prostate cancer: a randomized phase II study. Cytotherapy 2017;19:500-13.

286. Glabman RA, Choyke PL, Sato N. Cancer-associated fibroblasts: tumorigenicity and targeting for cancer therapy. Cancers 2022;14:3906.

287. Boudewijns S, Westdorp H, Koornstra RH, et al. Immune-related adverse events of dendritic cell vaccination correlate with immunologic and clinical outcome in stage III and IV melanoma patients. J Immunother 2016;39:241-8.

288. Agrawal A, Sridharan A, Prakash S, Agrawal H. Dendritic cells and aging: consequences for autoimmunity. Expert Rev Clin Immunol 2012;8:73-80.

Cite This Article

Export citation file: BibTeX | EndNote | RIS

OAE Style

Thomas R, Jerome JM, Krieger KL, Ashraf N, Rowley DR. The reactive stroma response regulates the immune landscape in prostate cancer. J Transl Genet Genom 2024;8:249-77. http://dx.doi.org/10.20517/jtgg.2024.15

AMA Style

Thomas R, Jerome JM, Krieger KL, Ashraf N, Rowley DR. The reactive stroma response regulates the immune landscape in prostate cancer. Journal of Translational Genetics and Genomics. 2024; 8(3): 249-77. http://dx.doi.org/10.20517/jtgg.2024.15

Chicago/Turabian Style

Rintu Thomas, John Michael Jerome, Kimiko L. Krieger, Naghmana Ashraf, David R. Rowley. 2024. "The reactive stroma response regulates the immune landscape in prostate cancer" Journal of Translational Genetics and Genomics. 8, no.3: 249-77. http://dx.doi.org/10.20517/jtgg.2024.15

ACS Style

Thomas, R.; Jerome JM.; Krieger KL.; Ashraf N.; Rowley DR. The reactive stroma response regulates the immune landscape in prostate cancer. J. Transl. Genet. Genom. 2024, 8, 249-77. http://dx.doi.org/10.20517/jtgg.2024.15

About This Article

Special Issue

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
78
Downloads
26
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Journal of Translational Genetics and Genomics
ISSN 2578-5281 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/