Next Article in Journal
The Effect of HMGB1 and HMGB2 on Transcriptional Regulation Differs in Neuroendocrine and Adenocarcinoma Models of Prostate Cancer
Previous Article in Journal
Microglial Transforming Growth Factor-β Signaling in Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Strategies for Overcoming Immune Evasion in Bladder Cancer

by
Juhyun Shin
1,
Jeong Won Park
1,
Seon Young Kim
1,
Jun Ho Lee
2,
Wahn Soo Choi
3 and
Hyuk Soon Kim
1,*
1
Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
2
Department of Korean Medicine, College of Korean Medicine, Woosuk University, Jeonju 54986, Republic of Korea
3
Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(6), 3105; https://doi.org/10.3390/ijms25063105
Submission received: 5 February 2024 / Revised: 4 March 2024 / Accepted: 5 March 2024 / Published: 7 March 2024

Abstract

:
Tumors intricately shape a highly immunosuppressive microenvironment, hampering effective antitumor immune responses through diverse mechanisms. Consequently, achieving optimal efficacy in cancer immunotherapy necessitates the reorganization of the tumor microenvironment and restoration of immune responses. Bladder cancer, ranking as the second most prevalent malignant tumor of the urinary tract, presents a formidable challenge. Immunotherapeutic interventions including intravesical BCG and immune checkpoint inhibitors such as atezolizumab, avelumab, and pembrolizumab have been implemented. However, a substantial unmet need persists as a majority of bladder cancer patients across all stages do not respond adequately to immunotherapy. Bladder cancer establishes a microenvironment that can actively hinder an efficient anti-tumor immune response. A deeper understanding of immune evasion mechanisms in bladder cancer will aid in suppressing recurrence and identifying viable therapeutic targets. This review seeks to elucidate mechanisms of immune evasion specific to bladder cancer and explore novel pathways and molecular targets that might circumvent resistance to immunotherapy.

1. Introduction

Bladder cancer is one of the most prevalent and malignant tumors that originates in cells within the bladder. In 2020, it ranked twelfth globally among diagnosed malignancies [1]. In its initial stages, bladder cancer may remain asymptomatic, while certain individuals may later experience symptoms such as urinary pain, the presence of blood in their urine, and increased frequency of urination. Upon diagnosis of bladder cancer, approximately 70% to 75% of patients exhibit non-muscle-invasive bladder cancer (NMIBC), while the remaining 25% to 30% manifest either muscle-invasive bladder cancer (MIBC) or distant metastases [2,3].
Bladder cancer intricately forms a highly immune-suppressive microenvironment, evading immune responses through diverse mechanisms. The bladder tumor microenvironment is characterized by a highly immune-suppressive milieu, including the accumulation of various types of immune cells with immunosuppressive phenotypes, such as regulatory T cells (Tregs), tolerogenic dendritic cells (tDCs), and tumor-associated macrophages (TAMs), along with an increased expression of programmed death-ligand 1 (PD-L1) and abnormal metabolism [4,5,6].
Therapeutic options for bladder cancer include chemotherapy and radical cystectomy for clinically localized cases, while systemic chemotherapy is available for patients with metastatic disease. Despite these aggressive treatment approaches, many patients do not respond to treatment or they experience recurrences, resulting in an unfavorable prognosis [7,8]. Due to immune suppression and tolerance caused by tumors, the clinical efficacy of cancer immunotherapy has been limited. Therefore, a deeper comprehension of immune evasion mechanisms in bladder cancer is necessary as it will play a crucial role in inhibiting recurrence and accurately identifying potential therapeutic targets.
Over the last 20 years, various groundbreaking immunotherapy strategies have been spotlighted in cancer treatment, such as PD-L1/PD-1 checkpoint inhibitors [9,10]. Nonetheless, there remains an unresolved challenge: most bladder cancer patients do not respond to immunotherapy across all stages of the disease. Therefore, there is a need to review candidate factors for overcoming resistance to immunotherapy in bladder cancer. This review aims to clarify how bladder cancer evades the immune system and investigates pathways and molecular targets that could overcome resistance to immunotherapy.

2. Bladder Cancer Therapy: Conventional Therapy and Immunotherapy

2.1. Standard Therapy of Bladder Cancer

Treating NMIBC offers a significant advantage over treating MIBC. This advantage lies in the ability to administer non-systemic chemotherapy directly into the bladder’s lumen, known as intravesical chemotherapy, thereby safeguarding normal body tissues from the potential toxicity of the agents used. Moreover, when combined with a transurethral resection of the bladder, intravesical chemotherapy has been proven to be highly effective in minimizing disease recurrence [11,12]. The most frequently utilized agents for this purpose include mitomycin C, epirubicin, thiotepa, gemcitabine, and doxorubicin. While these substances demonstrate effectiveness, there is still potential for enhancing their efficacy [13]. To improve efficacy, researchers have aimed to increase the absorption of chemotherapy by cancer cells following intravesical application. For instance, the utilization of albumin-bound nanoparticles to transport rapamycin or paclitaxel has been explored in bladder cancer treatment [14,15]. Preliminary animal studies have demonstrated that administering paclitaxel encased in polymers can reduce tumor weight [16]. Currently, the established treatment for MIBC involves the administration of neoadjuvant chemotherapy (NAC) before radical cystectomy (RC) [17,18,19]. Even with RC and NAC, 50% of patients still experience the development of distant metastases [20,21]. Additionally, adverse reactions to chemotherapy can limit NAC use among most patients [21]. Therefore, new treatments tailored for bladder cancer patients need to be developed.

2.2. Immunotherapy of Bladder Cancer

Presently, intravesical Bacillus Calmette-Guérin (BCG) therapy remains the foremost adjuvant therapy for high-risk NMIBC [22]. BCG, originally a tuberculosis (TB) vaccine, comprises live attenuated Mycobacterium tuberculosis. During the initiation phase of an immune response, BCG functions as a pathogen-associated molecular pattern (PAMP), stimulating pattern recognition receptors (PRRs) present on diverse cells, including bladder tumor cells and antigen-presenting cells (APCs), such as macrophages, dendritic cells (DCs), and others [8,23].
Most visible NMIBC can be removed through an endoscopic surgery known as transurethral resection (TUR). To address the high recurrence rates of NMIBC, intravesical therapies such as BCG can be utilized after TUR [7,24]. While BCG has remained a primary immunotherapy for many years, a considerable proportion of patients with NMIBC eventually transition to a state in which they do not respond effectively to BCG treatment [25,26]. Patients who do not achieve a complete response after BCG induction experience reduced five-year survival rates. Moreover, additional BCG therapy has been found to be ineffective [11,27,28]. BCG is used worldwide because certain chemotherapy options, such as mitomycin-C, may not be available in all countries [29]. Up to 35% of bladder cancer patients are treated with intravesical BCG, but 40 to 60% will have a recurrence of the tumor within 2 years [30].
Recent studies have demonstrated that immune checkpoints may play a role in the mechanism of BCG resistance [31]. The role of immune checkpoints involves preserving self-tolerance and regulating the magnitude and duration of inflammatory responses. However, their normal function in regulating this immune homeostasis is induced in cancer cells to evade immune attacks. Multiple antibodies that can block immune checkpoints have been approved for treating melanoma, lung cancer, and renal carcinoma, targeting additional immune checkpoints clinically [32]. In the ongoing randomized clinical trial KEYNOTE-057, pembrolizumab, a PD-1 inhibitor, has shown promising results in treating high-risk BCG-unresponsive NMIBC, leading to its recent FDA approval [33,34,35]. In a cohort of 96 patients, Balar et al. reported that an intravenous injection of pembrolizumab alone resulted in complete remission (CR) in 39 cases, but among them, 51% exhibited recurrent disease [36]. And recently, Atezolizumab, a monoclonal antibody that binds to PD-L1, was approved for the treatment of patients with MIBC [37]. However, there is a limitation that only MIBC patients who are not suitable for chemotherapy are included and there is no official maintenance treatment yet [37]. Therefore, additional evidence is needed to establish the safety and efficacy of immunotherapy in the treatment of bladder cancer and new therapeutic strategies are needed.

3. Factors That Bladder Tumors Exploit to Avoid Immune Responses

3.1. Immune Checkpoint Molecules (PD-1/PD-L1/CTLA-4)

Enhancing immunotherapies for bladder cancer will require deeper insights into the tumor microenvironment (TME) and effective techniques for leveraging the immune system against tumor progression. Immune checkpoints play a role in maintaining self-tolerance and controlling the intensity and duration of inflammatory reactions. However, tumors can exploit these pathways to evade the body’s immune response. In the past decade, immune checkpoint therapy has significantly transformed the treatment approach for bladder cancer [38,39,40]. Immune checkpoint therapy can stimulate an anti-tumor response by restoring T cell function through complex interactions among immune checkpoint molecules, such as PD-1, its ligand (PD-L1), and cytotoxic T lymphocyte-associated protein (CTLA-4) [32].
Expressed in activated T lymphocytes, PD-1 can regulate T cell effector functions and serve as a brake on T cell immune responses. Binding PD-1 to its counterpart, PD-L1, can lead to T cell exhaustion, a process that can reduce T cell activity (Figure 1). This is considered a mechanism to prevent auto-immunity [41]. The discovery of the involvement of the PD-1/PD-L1 axis in regulating T cells has led to preclinical studies demonstrating the overexpression of PD-1/PD-L1 in malignant cells [42]. Subsequently, PD-1/PD-L1 expression has been found to be higher in tumor tissue samples of bladder cancer patients than in normal tissues [41,42,43,44]. Patients unresponsive to BCG treatment show higher PD-L1 expression in both tumor and immune cells than those who are responsive to BCG treatment [45]. RNA sequencing analyses have revealed higher baseline PD-L1 levels in patients unresponsive to BCG [31,45]. The overall PD-1/PD-L1 pathway appears to be associated with localized bladder cancer immune response, indicating its value as a potential therapeutic target.
Expressed on activated T cells, CTLA-4 is a surface molecule that can interact with the B7 ligand found on B lymphocytes, DCs, and macrophages. While its full mechanism remains unclear, this element can exert a negative influence on the immune response. One proposed explanation is its structural similarity to CD28, suggesting that it may compete with CD28 for binding to ligands [46,47,48]. Other studies have shown that CTLA-4 expression is particularly high in MIBC and correlated with tumor size [49]. Currently, in addition to the already approved pembrolizumab (anti-PD-1), clinical trials for BCG non-responsive patients are ongoing. Typically, durvalumab is a human monoclonal IgG kappa antibody against PD-L1 and does not cause antibody-dependent cytotoxicity [50]. Clinical trials are being conducted on its efficacy in NMIBC patients, and it has been shown to be safe in combination with BCG [51]. There is also an ongoing phase I/II RIDEAU study aimed at determining whether systemic durvalumab in combination with anti-CTLA-4 tremelimumab is effective in patients with NMIBC [52]. The maximum tolerated dose (MTD) of durvalumab and 1-year HG relapse-free rate are the primary endpoints. There are no exact success figures for the ongoing combination treatment clinical trials because they are ongoing.

3.2. Human Leukocyte Antigen G (HLA-G)

HLA-G belongs to a non-traditional group of HLA class I molecules. It possesses immune-regulating characteristics [53]. HLA-G was initially reported to play crucial roles in maternal–fetal tolerance and tissue transplantation [54]. Since then, it has emerged as a significant immune checkpoint in various cancers, including bladder cancer, stomach cancer, colon cancer, and breast cancer [55,56,57,58]. Its inhibitory effect surpasses those of other immune checkpoints, as it can directly interact with key receptors, immunoglobulin-like transcript 2 (ILT2) and ILT4, thereby interfering with multiple stages of antitumor responses [59]. ILT2 exhibits distinct expression patterns on natural killer (NK) cells, T cells, B cells, monocytes, and DCs, while ILT4 is specific to myeloid cells [60,61] (Figure 1). Consequently, HLA-G interactions have the potential to disrupt initial or subsequent stages of immune responses [60]. Notably, HLA-G-induced suppressor cells such as Tregs and tDCs can perform long-term immunoregulatory functions, potentially blocking immunity [61]. Additionally, previous studies have demonstrated that HLA-G can promote cancer metastasis by inducing angiogenesis [62,63]. The expression of HLA-G in bladder cancer strongly correlates with tumor aggressiveness and poor survival outcomes [55].

3.3. Toll-like Receptor (TLR) Polyimorphism

TLRs are pattern recognition receptors (PRRs) that detect pathogen-associated molecular patterns (PAMPs) and initiate immune response. Upon TLR activation, NF-κB translocates to the nucleus and initiates the transcription of genes associated with cell proliferation, including cyclin D1, cyclin D2, c-Myc, and BCL-2 [64]. TLRs are expressed on APCs, such as macrophages and DCs, as well as on tumor cells [65,66]. The binding of ligands to TLRs induces APC maturation and the release of cytokines (TNF-α, IL-6, IL-12, and IFN) [65]. This is followed by the activation of NK cells and cytotoxic T cells, ultimately leading to the death of tumor cells [65]. However, TLR expression on bladder cancer cells can contribute to tumor growth and immune evasion by upregulating the growth factors and anti-apoptotic protein production [65,67]. Effective options achieved through TLR modulation may include blocking TLR involvement in inflammatory diseases and recruiting TLR signaling pathways against cancer.
The association between TLR polymorphisms and genetic susceptibility to various types of cancer has been extensively studied [68]. Hundreds of single-nucleotide polymorphisms (SNPs) have been identified in TLRs, but their functional consequences remain largely unknown. Nevertheless, several correlations between TLR polymorphisms and cancer have been reported [68,69]. Understanding TLR polymorphisms and their role in cancer will be helpful in selecting appropriate treatments. For example, the prevalence of TLR4 +3725GC and CC genotypes was found to be significantly increased in bladder cancer cases compared to controls [70]. TLR polymorphisms could potentially serve as therapeutic targets and prognostic indicators in bladder cancer.

3.4. Lactate

Cancer cells rely on aerobic glycolysis, known as the Warburg effect, to sustain growth even under normal oxygen conditions [71]. Several studies have demonstrated that increased lactate levels within the TME due to metabolic changes create an immunosuppressive environment [72,73]. Lactate dehydrogenase-A (LDH-A) plays a crucial role as an enzyme responsible for the final step of glycolysis, converting pyruvate into lactate. Elevated levels of LDH-A have been observed in various cancer types, such as cervical cancer and renal cell carcinoma [74], showing a strong association with cancer progression, metastasis, recurrence, and unfavorable clinical outcomes [74,75]. Lactate produced metabolically by LDH-A is considered an oncometabolite. Increased lactate levels in cancer are linked to a higher incidence of metastases and decreased survival [76]. Recent studies suggest that lactate contributes to the creation of an immunosuppressive TME by reducing the presence of NK cells and cytotoxic CD8+ T cells while suppressing the expression of interferon-γ (IFN-γ) [73] (Figure 1). Additionally, it has been reported that LDH-A-derived lactate within the TME can upregulate the expression of PD-L1 in lung cancer [77]. In bladder cancer, the overexpression of LDH-A has been correlated with tumor development and an unfavorable prognosis of patients [78]. Therefore, regulating LDH-A/lactate may be an effective approach to enhance the efficacy of anti-PD-1 therapy.

3.5. Prostaglandin E2 (PGE2)

PGE2, a bioactive lipid mediator, significantly contributes to immune evasion within the TME [79,80]. It interferes with anti-tumor immune responses by affecting various immune cells, including T cells [81], DCs [82,83], macrophages [84,85], and NK cells [86,87] (Figure 1). Regarding T cells, PGE2 can suppress the activation and proliferation of cytotoxic T cells and foster an immunosuppressive environment by inducing the expansion of Tregs [88,89,90]. Additionally, PGE2 can promote immune tolerance and tumorigenesis by inducing the polarization of macrophages towards the M2 phenotype [91]. PGE2 can also exert molecular effects by inhibiting effector T cell function and upregulating immune checkpoint molecules such as PD-L1 [92]. Increased levels of cyclooxygenase-2 (COX2), an essential enzyme in the biosynthetic pathway of PGE2, have been reported in various cancer types, including bladder cancer [93]. Recent studies suggest that COX2/PGE2 signaling plays a crucial role in the proliferation and regeneration of bladder cancer cells [94]. Considering these findings, appropriately regulating the metabolic process of PGE2 in bladder cancer could prove beneficial for immune-checkpoint-mediated treatment.

3.6. Tolerogenic Dendritic Cells (tDCs)

The TME encompasses various cell types collaborating in complex mechanisms, allowing cancer cells to evade immune surveillance. In addition to inhibition mediated by molecules such as PD-1 and CTLA-4, myeloid lineage cells can form a network of immunosuppressive cells, hindering the development of antitumor immunity [6]. This network includes TAMs and DCs. DCs primarily function to capture and present antigens, initiating the activation of CD4+ and CD8+ T cells. For the effective activation of T cells, DCs undergo a maturation process that involves the increased expression of antigen-presenting molecules (MHC class Ⅰ and Ⅱ), the upregulation of costimulatory molecules such as CD80 and CD86, and the production of immune-stimulating cytokine [95]. However, tumor cells typically employ various immunosuppressive strategies in their microenvironment to interfere with this process. Bladder cancer can influence the differentiation/maturation process of DCs, preventing the upregulation of co-stimulatory molecules and converting them into tDCs [96]. Recent studies have demonstrated that DCs exposed to Sialyl Tn (STn)-expressing bladder cancer cells show a decreased capacity to stimulate and guide T cells toward Th1 phenotypes, compromising their ability to generate an effective anti-tumor response [96,97,98,99].

3.7. Tumor-Associated Macrophages (TAMs)

TAMs exhibit diverse functions within the TME. The M1 polarization of TAMs can produce pro-inflammatory cytokines known to activate cytotoxic T cells and induce tumor immune responses. Conversely, M2 polarization can selectively attract regulatory T cells (Tregs) and Th2 cells while suppressing T cell responses [100]. IL-10-positive TAMs display an immunosuppressive phenotype similar to M2 macrophages in the context of MIBC, infiltrating the TME and suppressing immune responses [101] (Figure 1). The release of IL-10 by TAMs additionally heightens the expression levels of various immune checkpoints and co-inhibitory receptors (such as PD-L1, CTLA-4, and TIM-3). Compositions of chemokines within the TME substantially shape the direction and characteristics of macrophages. Specifically, chemokine (C-C motif) ligand 2 (CCL2) plays a crucial role in recruiting TAMs and contributes to bladder cancer cell proliferation, metastasis, and the establishment of an immunosuppressive environment in the TME [102,103]. Infiltrating TAMs can promote tumor development by altering the microenvironment of bladder cancer through the action of C-X-C motif chemokine ligand 8 (CXCL8) [103]. Conversely, CXCL8 has been shown to trigger pro-angiogenic and anti-apoptotic pathways when it is secreted by tumor cells [104,105,106].

3.8. Regulatory T Cells (Tregs)

Tregs play a crucial role in aiding tumors to evade anti-tumor immune responses and circumvent therapeutic interventions across various types of cancers [107,108]. A previous study has shown a marked increase in Tregs levels in peripheral blood samples of patients with bladder cancer [97]. In the same vein, the infiltration of T cell subpopulations within the tumor tissue showed significant reductions in tissue CD3+, CD4+, and CD8+ cells [109,110]. Patients with MIBC expressing forkhead box P3 (Foxp3), a transcription factor for Tregs, have a lower survival rate than patients with Foxp3-negative cancers [110]. Quinn et al. (2008) analyzed the effect of Treg inactivation before BCG vaccination on the development of protective immunity in a mouse model [111]. Although Treg inactivation increased the number of IL-2- or IFN-γ-producing CD4+ lymphocytes, the presence of CD4+CD25+ Tregs did not significantly affect the protective efficacy of the BCG vaccine [111]. However, the simultaneous suppression of Th2 cells and Tregs has been shown to increase Th1 responses, enhancing host protective immunity and BCG-induced vaccine efficacy [112]. Although our understanding of the impact of Tregs on bladder cancer remains incomplete, current efforts focus on tumor-infiltrating Tregs as a strategy for cancer therapy.

3.9. Cancer-Associated Fibroblasts (CAFs)

CAFs can produce key growth factors and cytokines as components of the ECM to promote tumor growth, metastasis, chemoresistance, and immune responses. Previous reports have revealed several mechanisms linking CAFs to immune evasion in cancer [113,114,115]. CAFs can remodel the extracellular matrix and secrete various factors such as cytokines, chemokines, and growth factors. These factors can influence the recruitment, function, and polarization of immune cells [116,117,118,119] and create an immunosuppressive environment that promotes tumor progression [120,121,122,123]. CAFs have been found to modulate immune checkpoint molecules such as PD-L1 [124,125]. The interaction between PD-L1 on CAFs and its receptor PD-1 on immune cells, such as T lymphocytes, can lead to immune suppression by inhibiting the activity of these immune cells, allowing tumors to evade immune attacks [126,127]. One study showed that a subset of CAFs, namely iCAFs, play an important role in promoting tumorigenesis or the migration of primary tumors by promoting angiogenesis and the epithelial–mesenchymal transition (EMT) [128]. Additionally, studies have confirmed that IL-6 is upregulated in bladder cancer iCAFs, indicating that the expression of IL-6R in bladder cancer cells is suitable for responding to the IL-6 cytokine secreted by iCAFs [129]. Siltuximab, an IL-6 blocking antibody, has already been approved by the FDA and has demonstrated antitumor efficacy against ovarian, prostate, and lung cancer [130]. Clinical trials investigating targeted approaches, such as IL-6 signaling inhibition in bladder cancer, are still lacking. However, this approach to disrupt CAF function may be a clinically promising strategy. C-X-C motif chemokine 12 (CXCL12) derived from CAFs has been established as one of the most potent chemokines that can regulate immune cells [131]. Understanding the intricate interplay between bladder cancer cells, CAFs, and the immune system is crucial for developing more effective therapeutic strategies.

4. Potential Therapeutic Strategies for Modulating Immune Evasion

4.1. Therapeutic Combination with Immune Checkpoint Inhibitor (ICI)

Currently, in the field of treating NMIBC, the main supporting therapy is intravesical BCG, but patients unresponsive to BCG undergo ICI treatment [132]. For patients diagnosed with MIBC, common treatment options include cisplatin chemotherapy or radical cystectomy. Nevertheless, there are instances in which patients are unsuitable for chemotherapy or choose not to undergo cystectomy [133]. In such cases, ICIs can serve as an alternative treatment method for MIBC patients [133]. Especially in the context of MIBC, ICI stands out as a significant treatment option due to its lower rates of recurrence and survival rates, even after cystectomy [134]. As of 2023, the FDA has approved three drugs (pembrolizumab, valrubicin, and nadofaragene firadenovec-vcng) for patients unresponsive to BCG and either ineligible for chemotherapy or refusing radical cystectomy [133].
Despite increased PD-L1 expression in bladder cancer patients, a significant number of patients do not respond to PD-1/PD-L1 inhibitors [135,136,137]. Therefore, the need for more accurate biomarkers is emphasized.
Combination therapy can be a strategy to enhance the efficacy of PD-1/PD-L1 inhibition (Figure 2). To date, the FDA has approved pembrolizumab only for patients with NMIBC and BCG failure who are unwilling or unable to undergo RC [138]. There is no approved combination administration method with ICI yet, but combination administration with BCG or ICI has shown effectiveness in clinical trials. Atezolizumab is a monoclonal antibody that inhibits the PD-L1/PD-1 pathway. In NCT02792192, 33.3% of the atezolizumab-only group and 41.7% of the atezolizumab + BCG group achieved complete remission (CR) [139]. The combination of CTLA-4 and PD-L1 inhibitors can result in heightened levels of CD4+ and CD8+ T cells, decreased Tregs, the elevated production of pro-inflammatory cytokines, and a shift in macrophage polarization from M2 to M1 [140,141]. The most recent NABUCCO clinical trial observed that in patients with stage Ⅲ bladder cancer who were not suitable for cisplatin therapy, the administration of preoperative ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) before RC resulted in a pathological complete response (pCR) rate of 46% [125,142].

4.2. Therapeutic Combination with HLA-G Target

In most cases, the HLA-G/ILT interaction can promote cancer cells to evade immune surveillance and anti-tumor immunity [143,144]. Therefore, a concurrent blockade of PD-1/PD-L1 and HLA-G/ILT2 might offer potential benefits in specific patient populations (Figure 2). Several in vitro studies have demonstrated that NK cells can more effectively eliminate HLA-G-negative leukemia, glioma, ovarian carcinoma, and hepatocellular carcinoma cell lines than those transfected with HLA-G [145,146,147,148]. HLA-G is frequently but heterogeneously expressed in various cancers, including bladder cancer. This expression pattern varies depending on tumor subtypes, tumor grades, tumor stages, and compositions of immune cell infiltration [149]. Therefore, further research is needed to explore the specific immunological functions of HLA-G and its potential therapeutic targeting capabilities. However, HLA-G has been proven to upregulate the expression of PD-1 on T lymphocytes. Thus, blocking HLA-G/ILT2-related PD-1/PD-L1 interactions might be an effective treatment strategy for bladder cancer [150].

4.3. TLR-Targeted Modulation

The expression and polymorphism of TLRs in bladder cancer cells are closely related to tumor development [65,70,151]. The activation of TLRs on tumor cells can trigger both pro- and anti- tumor responses, and the use of TLR antagonists and agonists has been discussed extensively [152,153,154,155]. Studies to prevent the immune evasion of cancer cells through TLR antagonists have been conducted in various carcinomas [156]. Indeed, significant tumor-destroying properties have been demonstrated for breast and ovarian carcinomas. [156]. However, some studies have reported more direct cytotoxic effects and in vivo anti-tumor effects of TLR agonists on bladder cancer cell lines compared to TLR antagonists [151,157].
Recent clinical studies on TLR agonists in cancer have shown promising results, especially when combined with other immunotherapies [158]. For instance, the combination of recombinant cancer vaccines with TLR3, 4, and 9 agonists has exhibited the ability to boost immune stimulation and enhance T cell responses in melanoma patients [159,160].
BCG, acting as a TLR2 and TLR4 agonist, remains a cornerstone in immunotherapy for NMIBC patients. However, alternative options are needed for patients unresponsive to BCG. Recently, imiquimod and TMX-202 (TLR7 agonists) were tested for bladder cancer immunotherapy, while CpG ODN (a TLR9 agonist) and HP-NAP (a TLR2 agonist) demonstrated reduced tumor growth in an MB49 bladder cancer mouse model [161,162,163,164]. As discussed earlier, genetic susceptibility to bladder cancer may vary depending on the TLR polymorphism. Studies indicate that variant alleles of TLR2 (ID+DD) are associated with disease susceptibility and risk of progression in bladder cancer [70]. Furthermore, the prevalence of TLR4 +3725G/C has been significantly increased in bladder cancer cases [165]. Therefore, these TLR polymorphisms could potentially serve as new therapeutic targets and prognostic indicators in bladder cancer.

4.4. LDH-A/lactate

Metabolites present within the TME significantly interfere with the function of immune cells, especially anti-tumor T cells. Therefore, therapeutic approaches targeting metabolic changes to improve the TME may provide promising complementary treatments in combination with ICI. According to a recent study, increased LDH-A in melanoma patients is correlated with a decreased response to anti-PD-1 immunotherapy [76]. In a mouse model of melanoma, blocking LDH-A can increase the production of IFN-γ and granzyme B in NK cells and CD8+ T cells [166]. Additionally, it can enhance anti-tumor immune response to immune checkpoint inhibitors [166]. Tumors with LDH-A deficiency exhibit notably higher levels of responsiveness to anti-PD-1 treatment than tumors expressing LDH-A [76]. This combination therapy can potently block the PD-1/PD-L1 pathway, thereby enhancing the infiltration and activation of NK cells and CD8+ T cells and increasing anti-tumor inflammatory responses (Figure 2).
LDH-A is upregulated in MIBC, promoting the proliferation of MIBC cells [78]. Therefore, it can be a viable option not only for treating NMIBC through combination treatment, but also for addressing MIBC, which generally has a less favorable prognosis.

4.5. mPGES1/15-PGDH

PGE2 contributes to tumor progression through a variety of mechanisms, including immunosuppression within the tumor environment and the proliferation or regeneration of cancer stem cells [92,167]. Elevated levels of COX2 have been documented in various cancers, including bladder cancer [93,168]. Recent studies have shown that inhibiting COX2 can reduce tumor-induced Treg activity and ultimately restore anti-tumor responses [169]. However, persistent COX2 inhibition can lead to adverse cardiovascular and gastrointestinal effects [170,171]. Therefore, targeting PGE2-producing enzyme microsomal prostaglandin E synthase-1 (mPGES1) and employing a targeted genetic enhancement of NAD+-dependent 15-hydroxyprostagladin dehydrogenase (15-PGDH), an enzyme that decomposes PGE2, might offer more efficient and safer approaches to combating cancer [92] (Figure 2). mPGES1 stands as the pivotal terminal enzyme implicated in the production of PGE2. Newer research has indicated that suppressing mPGES1 activity can lead to decreased PD-L1 expression in myeloid cells that infiltrate bladder tumor tissues [92]. Therefore, focusing on PGE2 metabolism may help reduce PD-L1-mediated immunosuppression.

4.6. Sialyl Tn (STn)

STn, a widespread tumor-associated carbohydrate antigen, is detected in more than 80% of human carcinomas but rarely found in normal tissues [172]. Within bladder cancer, STn is associated with the malignancy of the disease by amplifying the mobility and invasive potential of cancer cells [99]. STn can interfere with the expression of MHC-class Ⅱ and co-stimulatory molecules within DCs, reducing their ability to display cancer-related antigens to T cells [96] (Figure 1). Consequently, DCs become unresponsive to subsequent activation signals. Additionally, it can establish a tolerogenic microenvironment that enables cancer to evade attacks from both innate and adaptive immunity by impairing effective anti-tumor responses of Th1 cells [96,99]. DCs are used in current anti-tumor cell vaccines. The common strategy is injecting tumor cell antigens into a patient’s DCs [173]. Hence, merging therapies that can induce Th1 responses with DC-based treatments targeting cancer cells bearing STn holds promise as a strategy to stimulate protective anti-tumor responses [96] (Figure 2).
A study has revealed higher STn expression in metastatic MIBC compared to normal tissue, indicating a robust connection between STn antigens and tumor metastasis. This insight offers crucial molecular information for targeting aggressive bladder cancer cells [174]. Although additional research involving a larger cohort of patients is necessary before translating this into clinical practice, it holds potential as a valuable target for treating MIBC. Given its poor prognosis and limited therapeutic options compared to NMIBC, further exploration is warranted.

4.7. Regulation of Immune Cells within the TME through Chemokine Control

Chemokines and their receptors are recognized as mediators of chronic inflammation. They are important factors contributing to the progression of cancer [175]. Chemokines are increasingly recognized for their involvement in attracting a variety of cell types into the complex environment of tumors and inducing the spread of cancer cells to new sites. These encompass infiltrating cells such as TAMs and CAFs [175,176,177]. Therefore, blocking chemokines in the TME could be a promising approach to enhancing the efficacy of immunotherapy. CXCL12, also known as stromal cell-derived factor 1 (SDF1), is a chemokine crucially expressed across various tissues to maintain homeostasis. Its involvement in the development of bladder cancer has been extensively documented and discussed [178]. Within the TME, interaction between CXCL12 and its primary receptor C-X-C chemokine receptor type 4 (CXCR4) triggers downstream signaling pathways, leading to various outcomes, including the proliferation and evasion of immune cells. In a CXCL12-dependent manner, CAFs can effectively reduce the migration of CD8+ T cells towards the stromal region adjacent to the tumor, consequently limiting the infiltration of CD8+ T cells around tumor cells [179]. CXCL12 has the ability to rapidly increase the expression of PD-L1 in cancer cells within a short timeframe [131]. Suppressing CXCL12 produced by CAFs can notably reduce the impact of CAFs on the expression of PD-L1 [131] (Figure 2). A comprehensive screening of CAFs will offer deeper insights into the development of bladder cancer. Directing therapeutic efforts towards CXCL12 derived from CAFs could potentially be a valuable strategy for treating bladder cancer.
CCL2, a significant chemokine crucial for macrophages, exhibits high expression within cancer cells. It is actively secreted by these cells [102]. Several research studies have shown that CCL2 can promote the M2 polarization of TAMs, consequently amplifying immunosuppressive properties within the TME [180,181]. It has been found that CCL2 overexpression in bladder cancer is due to the FAM171B protein, which can act as an immune regulator and upregulate CCL2 expression [102]. Altering the immune microenvironment within tumors by targeting FAM171B could potentially serve as a viable strategy to increase the efficacy of immunotherapy when managing bladder cancer (Figure 2). Additionally, potential therapeutic approaches for bladder cancer involve the targeting of the CCL2-CCR2 axis, which could have encouraging results (Figure 2).
The CXCL8/CXCR2 pathway is crucial in human cancer. It contributes to tumor progression through various mechanisms [182]. Bladder cancer patients show notable increases in TAM infiltration than patients in a normal control group. Moreover, TAM levels show positive correlations with the expression of CXCL8 in bladder cancer tissues [104,183]. CXCL8 produced by TAMs has the potential to induce the expression of VEGF in bladder cancer cells. This stimulation contributes to changes in bladder cancer cell migration and invasion, consequently promoting bladder cancer progression [183]. Moreover, the engagement between bladder cancer cells and endothelial cells (ECs) can augment EC recruitment via the CXCL8–CXCR2 pathway [184]. Vascular ECs can interact with bladder cancer cells and tissues to promote cancer progression by activating the epidermal growth factor receptor (EGFR) signaling pathway [184]. This pathway can induce cancer proliferation, migration, and the secretion of CXC chemokines from bladder cancer cells. Therefore, targeting CXCL8 signaling presents a potential and promising therapeutic strategy for managing bladder cancer.

5. Conclusions

Immunotherapy for cancer has garnered significant attention as an exceedingly effective and promising approach. Despite notable progress in the development, clinical trials, and validation of therapeutics such as immune checkpoint inhibitors, gene-manipulated immune cells, and novel cancer vaccines, the clinical efficacy of cancer immunotherapy remains limited. The TME consists of a variety of cells that collaborate to assist cancer cells in evading immune surveillance and surviving therapeutic interventions. Within the TME, receptor–ligand interactions, metabolic byproducts, and the genetic sensitivity of these molecules can closely interact. Therefore, concurrently addressing factors involved in tumor-induced immune suppression during immunotherapy administration has the potential to further amplify anti-tumor immune responses. New insights into immune evasion in bladder cancer, as discussed earlier, suggest the potential to enhance cancer immunotherapy by targeting specific immune suppressive elements within the TME.

Author Contributions

H.S.K. contributed to study’s conception. J.S., J.W.P. and S.Y.K. performed the literature review and analysis. J.S. and H.S.K. wrote the draft of the manuscript. J.H.L. and W.S.C. provided intellectual input and wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Dong-A University research fund.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cao, W.; Chen, H.D.; Yu, Y.W.; Li, N.; Chen, W.Q. Changing profiles of cancer burden worldwide and in China: A secondary analysis of the global cancer statistics 2020. Chin. Med. J. 2021, 134, 783–791. [Google Scholar] [CrossRef]
  2. Claps, F.; van de Kamp, M.W.; Mayr, R.; Bostrom, P.J.; Shariat, S.F.; Hippe, K.; Bertz, S.; Neuzillet, Y.; Sanders, J.; Otto, W.; et al. Prognostic impact of variant histologies in urothelial bladder cancer treated with radical cystectomy. BJU Int. 2023, 132, 170–180. [Google Scholar] [CrossRef] [PubMed]
  3. Seidl, C. Targets for Therapy of Bladder Cancer. Semin. Nucl. Med. 2020, 50, 162–170. [Google Scholar] [CrossRef] [PubMed]
  4. Allegrezza, M.J.; Conejo-Garcia, J.R. Targeted Therapy and Immunosuppression in the Tumor Microenvironment. Trends Cancer 2017, 3, 19–27. [Google Scholar] [CrossRef] [PubMed]
  5. Kusmartsev, S.; Gabrilovich, D.I. Effect of tumor-derived cytokines and growth factors on differentiation and immune suppressive features of myeloid cells in cancer. Cancer Metastasis Rev. 2006, 25, 323–331. [Google Scholar] [CrossRef] [PubMed]
  6. Ostrand-Rosenberg, S.; Sinha, P.; Beury, D.W.; Clements, V.K. Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression. Semin. Cancer Biol. 2012, 22, 275–281. [Google Scholar] [CrossRef] [PubMed]
  7. Kawai, K.; Miyazaki, J.; Joraku, A.; Nishiyama, H.; Akaza, H. Bacillus Calmette-Guerin (BCG) immunotherapy for bladder cancer: Current understanding and perspectives on engineered BCG vaccine. Cancer Sci. 2013, 104, 22–27. [Google Scholar] [CrossRef] [PubMed]
  8. Han, J.; Gu, X.; Li, Y.; Wu, Q. Mechanisms of BCG in the treatment of bladder cancer-current understanding and the prospect. Biomed. Pharmacother. 2020, 129, 110393. [Google Scholar] [CrossRef]
  9. Okazaki, T.; Honjo, T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006, 27, 195–201. [Google Scholar] [CrossRef]
  10. Stenehjem, D.D.; Tran, D.; Nkrumah, M.A.; Gupta, S. PD1/PDL1 inhibitors for the treatment of advanced urothelial bladder cancer. Onco Targets Ther. 2018, 11, 5973–5989. [Google Scholar] [CrossRef]
  11. Babjuk, M.; Burger, M.; Comperat, E.M.; Gontero, P.; Mostafid, A.H.; Palou, J.; van Rhijn, B.W.G.; Roupret, M.; Shariat, S.F.; Sylvester, R.; et al. European Association of Urology Guidelines on Non-muscle-invasive Bladder Cancer (TaT1 and Carcinoma In Situ)—2019 Update. Eur. Urol. 2019, 76, 639–657. [Google Scholar] [CrossRef]
  12. Messing, E.M.; Tangen, C.M.; Lerner, S.P.; Sahasrabudhe, D.M.; Koppie, T.M.; Wood, D.P., Jr.; Mack, P.C.; Svatek, R.S.; Evans, C.P.; Hafez, K.S.; et al. Effect of Intravesical Instillation of Gemcitabine vs Saline Immediately Following Resection of Suspected Low-Grade Non-Muscle-Invasive Bladder Cancer on Tumor Recurrence: SWOG S0337 Randomized Clinical Trial. JAMA 2018, 319, 1880–1888. [Google Scholar] [CrossRef]
  13. Tran, L.; Xiao, J.F.; Agarwal, N.; Duex, J.E.; Theodorescu, D. Advances in bladder cancer biology and therapy. Nat. Rev. Cancer 2021, 21, 104–121. [Google Scholar] [CrossRef] [PubMed]
  14. Seager, C.M.; Puzio-Kuter, A.M.; Patel, T.; Jain, S.; Cordon-Cardo, C.; Mc Kiernan, J.; Abate-Shen, C. Intravesical delivery of rapamycin suppresses tumorigenesis in a mouse model of progressive bladder cancer. Cancer Prev. Res. 2009, 2, 1008–1014. [Google Scholar] [CrossRef] [PubMed]
  15. McKiernan, J.M.; Holder, D.D.; Ghandour, R.A.; Barlow, L.J.; Ahn, J.J.; Kates, M.; Badalato, G.M.; Roychoudhury, A.; Decastro, G.J.; Benson, M.C. Phase II trial of intravesical nanoparticle albumin bound paclitaxel for the treatment of nonmuscle invasive urothelial carcinoma of the bladder after bacillus Calmette-Guerin treatment failure. J. Urol. 2014, 192, 1633–1638. [Google Scholar] [CrossRef] [PubMed]
  16. Tamura, K.; Kikuchi, E.; Konno, T.; Ishihara, K.; Matsumoto, K.; Miyajima, A.; Oya, M. Therapeutic effect of intravesical administration of paclitaxel solubilized with poly(2-methacryloyloxyethyl phosphorylcholine-co-n-butyl methacrylate) in an orthotopic bladder cancer model. BMC Cancer 2015, 15, 317. [Google Scholar] [CrossRef]
  17. Witjes, J.A.; Bruins, H.M.; Cathomas, R.; Comperat, E.M.; Cowan, N.C.; Gakis, G.; Hernandez, V.; Linares Espinos, E.; Lorch, A.; Neuzillet, Y.; et al. European Association of Urology Guidelines on Muscle-invasive and Metastatic Bladder Cancer: Summary of the 2020 Guidelines. Eur. Urol. 2021, 79, 82–104. [Google Scholar] [CrossRef] [PubMed]
  18. Hermans, T.J.N.; Voskuilen, C.S.; van der Heijden, M.S.; Schmitz-Drager, B.J.; Kassouf, W.; Seiler, R.; Kamat, A.M.; Grivas, P.; Kiltie, A.E.; Black, P.C.; et al. Neoadjuvant treatment for muscle-invasive bladder cancer: The past, the present, and the future. Urol. Oncol. 2018, 36, 413–422. [Google Scholar] [CrossRef] [PubMed]
  19. Hu, J.; Chen, J.; Ou, Z.; Chen, H.; Liu, Z.; Chen, M.; Zhang, R.; Yu, A.; Cao, R.; Zhang, E.; et al. Neoadjuvant immunotherapy, chemotherapy, and combination therapy in muscle-invasive bladder cancer: A multi-center real-world retrospective study. Cell Rep. Med. 2022, 3, 100785. [Google Scholar] [CrossRef]
  20. International Collaboration of Trialists; Medical Research Council Advanced Bladder Cancer Working Party (now the National Cancer Research Institute Bladder Cancer Clinical Studies Group); European Organisation for Research and Treatment of Cancer Genito-Urinary Tract Cancer Group; Australian Bladder Cancer Study Group; National Cancer Institute of Canada Clinical Trials Group; Finnbladder; Norwegian Bladder Cancer Study Group; Club Urologico Espanol de Tratamiento Oncologico Group; Griffiths, G.; Hall, R.; et al. International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: Long-term results of the BA06 30894 trial. J. Clin. Oncol. 2011, 29, 2171–2177. [Google Scholar]
  21. Raj, G.V.; Karavadia, S.; Schlomer, B.; Arriaga, Y.; Lotan, Y.; Sagalowsky, A.; Frenkel, E. Contemporary use of perioperative cisplatin-based chemotherapy in patients with muscle-invasive bladder cancer. Cancer 2011, 117, 276–282. [Google Scholar] [CrossRef]
  22. Pettenati, C.; Ingersoll, M.A. Mechanisms of BCG immunotherapy and its outlook for bladder cancer. Nat. Rev. Urol. 2018, 15, 615–625. [Google Scholar] [CrossRef] [PubMed]
  23. Moriwaki, Y.; Begum, N.A.; Kobayashi, M.; Matsumoto, M.; Toyoshima, K.; Seya, T. Mycobacterium bovis Bacillus Calmette-Guerin and its cell wall complex induce a novel lysosomal membrane protein, SIMPLE, that bridges the missing link between lipopolysaccharide and p53-inducible gene, LITAF(PIG7), and estrogen-inducible gene, EET-1. J. Biol. Chem. 2001, 276, 23065–23076. [Google Scholar] [CrossRef]
  24. Comperat, E.; Amin, M.B.; Cathomas, R.; Choudhury, A.; De Santis, M.; Kamat, A.; Stenzl, A.; Thoeny, H.C.; Witjes, J.A. Current best practice for bladder cancer: A narrative review of diagnostics and treatments. Lancet 2022, 400, 1712–1721. [Google Scholar] [CrossRef]
  25. Kamat, A.M.; Colombel, M.; Sundi, D.; Lamm, D.; Boehle, A.; Brausi, M.; Buckley, R.; Persad, R.; Palou, J.; Soloway, M.; et al. BCG-unresponsive non-muscle-invasive bladder cancer: Recommendations from the IBCG. Nat. Rev. Urol. 2017, 14, 244–255. [Google Scholar] [CrossRef]
  26. Duplisea, J.J.; Mokkapati, S.; Plote, D.; Schluns, K.S.; McConkey, D.J.; Yla-Herttuala, S.; Parker, N.R.; Dinney, C.P. The development of interferon-based gene therapy for BCG unresponsive bladder cancer: From bench to bedside. World J. Urol. 2019, 37, 2041–2049. [Google Scholar] [CrossRef] [PubMed]
  27. Lerner, S.P.; Tangen, C.M.; Sucharew, H.; Wood, D.; Crawford, E.D. Failure to achieve a complete response to induction BCG therapy is associated with increased risk of disease worsening and death in patients with high risk non-muscle invasive bladder cancer. Urol. Oncol. 2009, 27, 155–159. [Google Scholar] [CrossRef]
  28. Chang, S.S.; Bochner, B.H.; Chou, R.; Dreicer, R.; Kamat, A.M.; Lerner, S.P.; Lotan, Y.; Meeks, J.J.; Michalski, J.M.; Morgan, T.M.; et al. Treatment of Non-Metastatic Muscle-Invasive Bladder Cancer: AUA/ASCO/ASTRO/SUO Guideline. J. Urol. 2017, 198, 552–559. [Google Scholar] [CrossRef] [PubMed]
  29. Wroclawski, M.L.; Schutz, F.A.; Cha, J.D.; Soares, A. Alternative Therapies to Bacillus Calmette-Guerin Shortage for Nonmuscle Invasive Bladder Cancer in Brazil and Other Underdeveloped Countries: Management Considerations. J. Glob. Oncol. 2019, 5, 1–9. [Google Scholar] [CrossRef]
  30. Kamat, A.M.; Sylvester, R.J.; Bohle, A.; Palou, J.; Lamm, D.L.; Brausi, M.; Soloway, M.; Persad, R.; Buckley, R.; Colombel, M.; et al. Definitions, End Points, and Clinical Trial Designs for Non-Muscle-Invasive Bladder Cancer: Recommendations from the International Bladder Cancer Group. J. Clin. Oncol. 2016, 34, 1935–1944. [Google Scholar] [CrossRef]
  31. Kates, M.; Matoso, A.; Choi, W.; Baras, A.S.; Daniels, M.J.; Lombardo, K.; Brant, A.; Mikkilineni, N.; McConkey, D.J.; Kamat, A.M.; et al. Adaptive Immune Resistance to Intravesical BCG in Non-Muscle Invasive Bladder Cancer: Implications for Prospective BCG-Unresponsive Trials. Clin. Cancer Res. 2020, 26, 882–891. [Google Scholar] [CrossRef] [PubMed]
  32. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer 2016, 16, 275–287. [Google Scholar] [CrossRef]
  33. Wright, K.M. FDA Approves Pembrolizumab for BCG-Unresponsive NMIBC. Oncology 2020, 34, 44. [Google Scholar] [PubMed]
  34. Claps, F.; Pavan, N.; Ongaro, L.; Tierno, D.; Grassi, G.; Trombetta, C.; Tulone, G.; Simonato, A.; Bartoletti, R.; Mertens, L.S.; et al. BCG-Unresponsive Non-Muscle-Invasive Bladder Cancer: Current Treatment Landscape and Novel Emerging Molecular Targets. Int. J. Mol. Sci. 2023, 24, 12596. [Google Scholar] [CrossRef] [PubMed]
  35. de Jong, F.C.; Rutten, V.C.; Zuiverloon, T.C.M.; Theodorescu, D. Improving Anti-PD-1/PD-L1 Therapy for Localized Bladder Cancer. Int. J. Mol. Sci. 2021, 22, 2800. [Google Scholar] [CrossRef]
  36. Balar, A.V.; Kamat, A.M.; Kulkarni, G.S.; Uchio, E.M.; Boormans, J.L.; Roumiguie, M.; Krieger, L.E.M.; Singer, E.A.; Bajorin, D.F.; Grivas, P.; et al. Pembrolizumab monotherapy for the treatment of high-risk non-muscle-invasive bladder cancer unresponsive to BCG (KEYNOTE-057): An open-label, single-arm, multicentre, phase 2 study. Lancet Oncol. 2021, 22, 919–930. [Google Scholar] [CrossRef]
  37. Suzman, D.L.; Agrawal, S.; Ning, Y.M.; Maher, V.E.; Fernandes, L.L.; Karuri, S.; Tang, S.; Sridhara, R.; Schroeder, J.; Goldberg, K.B.; et al. FDA Approval Summary: Atezolizumab or Pembrolizumab for the Treatment of Patients with Advanced Urothelial Carcinoma Ineligible for Cisplatin-Containing Chemotherapy. Oncologist 2019, 24, 563–569. [Google Scholar] [CrossRef]
  38. Sharma, P.; Allison, J.P. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell 2015, 161, 205–214. [Google Scholar] [CrossRef]
  39. Schulz, G.B.; Todorova, R.; Braunschweig, T.; Rodler, S.; Volz, Y.; Eismann, L.; Pfitzinger, P.; Jokisch, F.; Buchner, A.; Stief, C.; et al. PD-L1 expression in bladder cancer: Which scoring algorithm in what tissue? Urol. Oncol. 2021, 39, 734.e1–734.e10. [Google Scholar] [CrossRef]
  40. Lopez-Beltran, A.; Cimadamore, A.; Blanca, A.; Massari, F.; Vau, N.; Scarpelli, M.; Cheng, L.; Montironi, R. Immune Checkpoint Inhibitors for the Treatment of Bladder Cancer. Cancers 2021, 13, 131. [Google Scholar] [CrossRef]
  41. Sharpe, A.H.; Pauken, K.E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 2018, 18, 153–167. [Google Scholar] [CrossRef] [PubMed]
  42. Zhou, T.C.; Sankin, A.I.; Porcelli, S.A.; Perlin, D.S.; Schoenberg, M.P.; Zang, X. A review of the PD-1/PD-L1 checkpoint in bladder cancer: From mediator of immune escape to target for treatment. Urol. Oncol. 2017, 35, 14–20. [Google Scholar] [CrossRef] [PubMed]
  43. Boorjian, S.A.; Sheinin, Y.; Crispen, P.L.; Farmer, S.A.; Lohse, C.M.; Kuntz, S.M.; Leibovich, B.C.; Kwon, E.D.; Frank, I. T-cell coregulatory molecule expression in urothelial cell carcinoma: Clinicopathologic correlations and association with survival. Clin. Cancer Res. 2008, 14, 4800–4808. [Google Scholar] [CrossRef] [PubMed]
  44. Inman, B.A.; Sebo, T.J.; Frigola, X.; Dong, H.; Bergstralh, E.J.; Frank, I.; Fradet, Y.; Lacombe, L.; Kwon, E.D. PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: Associations with localized stage progression. Cancer 2007, 109, 1499–1505. [Google Scholar] [CrossRef] [PubMed]
  45. Pierconti, F.; Raspollini, M.R.; Martini, M.; Larocca, L.M.; Bassi, P.F.; Bientinesi, R.; Baroni, G.; Minervini, A.; Petracco, G.; Pini, G.M.; et al. PD-L1 expression in bladder primary in situ urothelial carcinoma: Evaluation in BCG-unresponsive patients and BCG responders. Virchows Arch. 2020, 477, 269–277. [Google Scholar] [CrossRef] [PubMed]
  46. Peggs, K.S.; Quezada, S.A.; Chambers, C.A.; Korman, A.J.; Allison, J.P. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J. Exp. Med. 2009, 206, 1717–1725. [Google Scholar] [CrossRef] [PubMed]
  47. van der Merwe, P.A.; Davis, S.J. Molecular interactions mediating T cell antigen recognition. Annu. Rev. Immunol. 2003, 21, 659–684. [Google Scholar] [CrossRef]
  48. Carreno, B.M.; Bennett, F.; Chau, T.A.; Ling, V.; Luxenberg, D.; Jussif, J.; Baroja, M.L.; Madrenas, J. CTLA-4 (CD152) can inhibit T cell activation by two different mechanisms depending on its level of cell surface expression. J. Immunol. 2000, 165, 1352–1356. [Google Scholar] [CrossRef]
  49. Zhang, W.; Shi, L.; Zhao, Z.; Du, P.; Ye, X.; Li, D.; Cai, Z.; Han, J.; Cai, J. Disruption of CTLA-4 expression on peripheral blood CD8 + T cell enhances anti-tumor efficacy in bladder cancer. Cancer Chemother. Pharmacol. 2019, 83, 911–920. [Google Scholar] [CrossRef]
  50. Hahn, N.M.; O’Donnell, M.A.; Efstathiou, J.A.; Zahurak, M.; Rosner, G.L.; Smith, J.; Kates, M.R.; Bivalacqua, T.J.; Tran, P.T.; Song, D.Y.; et al. A Phase 1 Trial of Durvalumab in Combination with Bacillus Calmette-Guerin (BCG) or External Beam Radiation Therapy in Patients with BCG-unresponsive Non-muscle-Invasive Bladder Cancer: The Hoosier Cancer Research Network GU16-243 ADAPT-BLADDER Study. Eur. Urol. 2023, 83, 486–494. [Google Scholar] [CrossRef]
  51. Hahn, N. PhAse 1/2 StuDy of Modern ImmunotherApy in BCG-Unresponsive, BCG-RelaPsing, and High-Risk BCG-Naive Non-muscle Invasive UroThelial Carcinoma of the BLADDER. 2023. Available online: https://clinicaltrials.gov/study/NCT03317158 (accessed on 22 August 2023).
  52. Black, P. Phase I/II Trial of Local Cystoscopic Injection of Tremelimumab Plus Systemic Durvalumab (MEDI4736) for High Risk Non-Muscle Invasive bladder Cancer. 2021. Available online: https://clinicaltrials.gov/study/NCT05120622 (accessed on 7 September 2023).
  53. Carosella, E.D.; Dausset, J.; Rouas-Freiss, N. Immunotolerant functions of HLA-G. Cell Mol. Life Sci. 1999, 55, 327–333. [Google Scholar] [CrossRef] [PubMed]
  54. Rouas-Freiss, N.; Goncalves, R.M.; Menier, C.; Dausset, J.; Carosella, E.D. Direct evidence to support the role of HLA-G in protecting the fetus from maternal uterine natural killer cytolysis. Proc. Natl. Acad. Sci. USA 1997, 94, 11520–11525. [Google Scholar] [CrossRef] [PubMed]
  55. Moavro, D.E.; Andrini, L.; Iribarne, A.; Colaci, P.; Garcia, M. HLA-G expression in transitional bladder carcinoma. Relationship with tumor invasion level and patient survival: Experience in a public hospital in Argentina. Hum. Immunol. 2023, 84, 423–427. [Google Scholar] [CrossRef] [PubMed]
  56. Wan, R.; Wang, Z.W.; Li, H.; Peng, X.D.; Liu, G.Y.; Ou, J.M.; Cheng, A.Q. Human Leukocyte Antigen-G Inhibits the Anti-Tumor Effect of Natural Killer Cells via Immunoglobulin-Like Transcript 2 in Gastric Cancer. Cell Physiol. Biochem. 2017, 44, 1828–1841. [Google Scholar] [CrossRef] [PubMed]
  57. Ye, S.R.; Yang, H.; Li, K.; Dong, D.D.; Lin, X.M.; Yie, S.M. Human leukocyte antigen G expression: As a significant prognostic indicator for patients with colorectal cancer. Mod. Pathol. 2007, 20, 375–383. [Google Scholar] [CrossRef] [PubMed]
  58. He, X.; Dong, D.D.; Yie, S.M.; Yang, H.; Cao, M.; Ye, S.R.; Li, K.; Liu, J.; Chen, J. HLA-G expression in human breast cancer: Implications for diagnosis and prognosis, and effect on allocytotoxic lymphocyte response after hormone treatment in vitro. Ann. Surg. Oncol. 2010, 17, 1459–1469. [Google Scholar] [CrossRef]
  59. Carosella, E.D.; Rouas-Freiss, N.; Tronik-Le Roux, D.; Moreau, P.; LeMaoult, J. HLA-G: An Immune Checkpoint Molecule. Adv. Immunol. 2015, 127, 33–144. [Google Scholar]
  60. Martin-Villa, J.M.; Vaquero-Yuste, C.; Molina-Alejandre, M.; Juarez, I.; Suarez-Trujillo, F.; Lopez-Nares, A.; Palacio-Gruber, J.; Barrera-Gutierrez, L.; Fernandez-Cruz, E.; Rodriguez-Sainz, C.; et al. HLA-G: Too Much or Too Little? Role in Cancer and Autoimmune Disease. Front. Immunol. 2022, 13, 796054. [Google Scholar] [CrossRef]
  61. Carosella, E.D.; HoWangYin, K.Y.; Favier, B.; LeMaoult, J. HLA-G-dependent suppressor cells: Diverse by nature, function, and significance. Hum. Immunol. 2008, 69, 700–707. [Google Scholar] [CrossRef]
  62. Raica, M.; Jitariu, A.A.; Cimpean, A.M. Lymphangiogenesis and Anti-lymphangiogenesis in Cutaneous Melanoma. Anticancer. Res. 2016, 36, 4427–4435. [Google Scholar] [CrossRef]
  63. Zhang, P.; Guo, X.; Li, J.; Yu, S.; Wang, L.; Jiang, G.; Yang, D.; Wei, Z.; Zhang, N.; Liu, J.; et al. Immunoglobulin-like transcript 4 promotes tumor progression and metastasis and up-regulates VEGF-C expression via ERK signaling pathway in non-small cell lung cancer. Oncotarget 2015, 6, 13550–13563. [Google Scholar] [CrossRef]
  64. Vallabhapurapu, S.; Karin, M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu. Rev. Immunol. 2009, 27, 693–733. [Google Scholar] [CrossRef]
  65. Ohadian Moghadam, S.; Nowroozi, M.R. Toll-like receptors: The role in bladder cancer development, progression and immunotherapy. Scand. J. Immunol. 2019, 90, e12818. [Google Scholar] [CrossRef]
  66. Afsharimoghaddam, A.; Soleimani, M.; Lashay, A.; Dehghani, M.; Sepehri, Z. Controversial roles played by toll like receptor 4 in urinary bladder cancer; A systematic review. Life Sci. 2016, 158, 31–36. [Google Scholar] [CrossRef] [PubMed]
  67. Sato, Y.; Goto, Y.; Narita, N.; Hoon, D.S. Cancer Cells Expressing Toll-like Receptors and the Tumor Microenvironment. Cancer Microenviron. 2009, 2 (Suppl. S1), 205–214. [Google Scholar] [CrossRef] [PubMed]
  68. El-Omar, E.M.; Ng, M.T.; Hold, G.L. Polymorphisms in Toll-like receptor genes and risk of cancer. Oncogene 2008, 27, 244–252. [Google Scholar] [CrossRef] [PubMed]
  69. Hold, G.L.; Rabkin, C.S.; Chow, W.H.; Smith, M.G.; Gammon, M.D.; Risch, H.A.; Vaughan, T.L.; McColl, K.E.; Lissowska, J.; Zatonski, W.; et al. A functional polymorphism of toll-like receptor 4 gene increases risk of gastric carcinoma and its precursors. Gastroenterology 2007, 132, 905–912. [Google Scholar] [CrossRef]
  70. Shen, Y.; Liu, Y.; Liu, S.; Zhang, A. Toll-like receptor 4 gene polymorphisms and susceptibility to bladder cancer. Pathol. Oncol. Res. 2013, 19, 275–280. [Google Scholar] [CrossRef] [PubMed]
  71. Zois, C.E.; Harris, A.L. Glycogen metabolism has a key role in the cancer microenvironment and provides new targets for cancer therapy. J. Mol. Med. 2016, 94, 137–154. [Google Scholar] [CrossRef] [PubMed]
  72. Romero-Garcia, S.; Moreno-Altamirano, M.M.; Prado-Garcia, H.; Sanchez-Garcia, F.J. Lactate Contribution to the Tumor Microenvironment: Mechanisms, Effects on Immune Cells and Therapeutic Relevance. Front. Immunol. 2016, 7, 52. [Google Scholar] [CrossRef]
  73. Scott, K.E.; Cleveland, J.L. Lactate Wreaks Havoc on Tumor-Infiltrating T and NK Cells. Cell Metab. 2016, 24, 649–650. [Google Scholar] [CrossRef] [PubMed]
  74. Girgis, H.; Masui, O.; White, N.M.; Scorilas, A.; Rotondo, F.; Seivwright, A.; Gabril, M.; Filter, E.R.; Girgis, A.H.; Bjarnason, G.A.; et al. Lactate dehydrogenase A is a potential prognostic marker in clear cell renal cell carcinoma. Mol. Cancer 2014, 13, 101. [Google Scholar] [CrossRef] [PubMed]
  75. Walenta, S.; Wetterling, M.; Lehrke, M.; Schwickert, G.; Sundfor, K.; Rofstad, E.K.; Mueller-Klieser, W. High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res. 2000, 60, 916–921. [Google Scholar] [PubMed]
  76. Daneshmandi, S.; Wegiel, B.; Seth, P. Blockade of Lactate Dehydrogenase-A (LDH-A) Improves Efficacy of Anti-Programmed Cell Death-1 (PD-1) Therapy in Melanoma. Cancers 2019, 11, 450. [Google Scholar] [CrossRef] [PubMed]
  77. Seth, P.; Csizmadia, E.; Hedblom, A.; Vuerich, M.; Xie, H.; Li, M.; Longhi, M.S.; Wegiel, B. Deletion of Lactate Dehydrogenase-A in Myeloid Cells Triggers Antitumor Immunity. Cancer Res. 2017, 77, 3632–3643. [Google Scholar] [CrossRef]
  78. Jiang, F.; Ma, S.; Xue, Y.; Hou, J.; Zhang, Y. LDH-A promotes malignant progression via activation of epithelial-to-mesenchymal transition and conferring stemness in muscle-invasive bladder cancer. Biochem. Biophys. Res. Commun. 2016, 469, 985–992. [Google Scholar] [CrossRef]
  79. Wei, J.; Zhang, J.; Wang, D.; Cen, B.; Lang, J.D.; DuBois, R.N. The COX-2-PGE2 Pathway Promotes Tumor Evasion in Colorectal Adenomas. Cancer Prev. Res. 2022, 15, 285–296. [Google Scholar] [CrossRef] [PubMed]
  80. Qiu, J.; Li, Q.; Li, J.; Zhou, F.; Sang, P.; Xia, Z.; Wang, W.; Wang, L.; Yu, Y.; Jiang, J. Complementary roles of EP2 and EP4 receptors in malignant glioma. Br. J. Pharmacol. 2023, 180, 2623–2640. [Google Scholar] [CrossRef]
  81. Wang, J.; Zhang, L.; Kang, D.; Yang, D.; Tang, Y. Activation of PGE2/EP2 and PGE2/EP4 signaling pathways positively regulate the level of PD-1 in infiltrating CD8(+) T cells in patients with lung cancer. Oncol. Lett. 2018, 15, 552–558. [Google Scholar] [CrossRef]
  82. Bayerl, F.; Meiser, P.; Donakonda, S.; Hirschberger, A.; Lacher, S.B.; Pedde, A.M.; Hermann, C.D.; Elewaut, A.; Knolle, M.; Ramsauer, L.; et al. Tumor-derived prostaglandin E2 programs cDC1 dysfunction to impair intratumoral orchestration of anti-cancer T cell responses. Immunity 2023, 56, 1341–1358 e11. [Google Scholar] [CrossRef]
  83. Harizi, H.; Juzan, M.; Pitard, V.; Moreau, J.F.; Gualde, N. Cyclooxygenase-2-issued prostaglandin e(2) enhances the production of endogenous IL-10, which down-regulates dendritic cell functions. J. Immunol. 2002, 168, 2255–2263. [Google Scholar] [CrossRef]
  84. Johansson, J.U.; Pradhan, S.; Lokteva, L.A.; Woodling, N.S.; Ko, N.; Brown, H.D.; Wang, Q.; Loh, C.; Cekanaviciute, E.; Buckwalter, M.; et al. Suppression of inflammation with conditional deletion of the prostaglandin E2 EP2 receptor in macrophages and brain microglia. J. Neurosci. 2013, 33, 16016–16032. [Google Scholar] [CrossRef]
  85. Akaogi, J.; Yamada, H.; Kuroda, Y.; Nacionales, D.C.; Reeves, W.H.; Satoh, M. Prostaglandin E2 receptors EP2 and EP4 are up-regulated in peritoneal macrophages and joints of pristane-treated mice and modulate TNF-alpha and IL-6 production. J. Leukoc. Biol. 2004, 76, 227–236. [Google Scholar] [CrossRef]
  86. Holt, D.; Ma, X.; Kundu, N.; Fulton, A. Prostaglandin E(2) (PGE (2)) suppresses natural killer cell function primarily through the PGE(2) receptor EP4. Cancer Immunol. Immunother. 2011, 60, 1577–1586. [Google Scholar] [CrossRef] [PubMed]
  87. Martinet, L.; Jean, C.; Dietrich, G.; Fournie, J.J.; Poupot, R. PGE2 inhibits natural killer and gamma delta T cell cytotoxicity triggered by NKR and TCR through a cAMP-mediated PKA type I-dependent signaling. Biochem. Pharmacol. 2010, 80, 838–845. [Google Scholar] [CrossRef]
  88. Thumkeo, D.; Punyawatthananukool, S.; Prasongtanakij, S.; Matsuura, R.; Arima, K.; Nie, H.; Yamamoto, R.; Aoyama, N.; Hamaguchi, H.; Sugahara, S.; et al. PGE(2)-EP2/EP4 signaling elicits immunosuppression by driving the mregDC-Treg axis in inflammatory tumor microenvironment. Cell Rep. 2022, 39, 110914. [Google Scholar] [CrossRef] [PubMed]
  89. Baratelli, F.; Lin, Y.; Zhu, L.; Yang, S.C.; Heuze-Vourc’h, N.; Zeng, G.; Reckamp, K.; Dohadwala, M.; Sharma, S.; Dubinett, S.M. Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells. J. Immunol. 2005, 175, 1483–1490. [Google Scholar] [CrossRef] [PubMed]
  90. Li, H.; Chen, H.Y.; Liu, W.X.; Jia, X.X.; Zhang, J.G.; Ma, C.L.; Zhang, X.J.; Yu, F.; Cong, B. Prostaglandin E(2) restrains human Treg cell differentiation via E prostanoid receptor 2-protein kinase A signaling. Immunol. Lett. 2017, 191, 63–72. [Google Scholar] [CrossRef] [PubMed]
  91. Dubey, P.; Shrivastava, R.; Tripathi, C.; Jain, N.K.; Tewari, B.N.; Lone, M.U.; Baghel, K.S.; Kumar, V.; Misra, S.; Bhadauria, S.; et al. Cyclooxygenase-2 inhibition attenuates hypoxic cancer cells induced m2-polarization of macrophages. Cell Mol. Biol. 2014, 60, 10–15. [Google Scholar] [PubMed]
  92. Prima, V.; Kaliberova, L.N.; Kaliberov, S.; Curiel, D.T.; Kusmartsev, S. COX2/mPGES1/PGE2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells. Proc. Natl. Acad. Sci. USA 2017, 114, 1117–1122. [Google Scholar] [CrossRef] [PubMed]
  93. Ristimaki, A.; Nieminen, O.; Saukkonen, K.; Hotakainen, K.; Nordling, S.; Haglund, C. Expression of cyclooxygenase-2 in human transitional cell carcinoma of the urinary bladder. Am. J. Pathol. 2001, 158, 849–853. [Google Scholar] [CrossRef]
  94. Liu, Q.; Yuan, W.; Tong, D.; Liu, G.; Lan, W.; Zhang, D.; Xiao, H.; Zhang, Y.; Huang, Z.; Yang, J.; et al. Metformin represses bladder cancer progression by inhibiting stem cell repopulation via COX2/PGE2/STAT3 axis. Oncotarget 2016, 7, 28235–28246. [Google Scholar] [CrossRef] [PubMed]
  95. Langenkamp, A.; Messi, M.; Lanzavecchia, A.; Sallusto, F. Kinetics of dendritic cell activation: Impact on priming of TH1, TH2 and nonpolarized T cells. Nat. Immunol. 2000, 1, 311–316. [Google Scholar] [CrossRef] [PubMed]
  96. Carrascal, M.A.; Severino, P.F.; Guadalupe Cabral, M.; Silva, M.; Ferreira, J.A.; Calais, F.; Quinto, H.; Pen, C.; Ligeiro, D.; Santos, L.L.; et al. Sialyl Tn-expressing bladder cancer cells induce a tolerogenic phenotype in innate and adaptive immune cells. Mol. Oncol. 2014, 8, 753–765. [Google Scholar] [CrossRef] [PubMed]
  97. Carlos, C.A.; Dong, H.F.; Howard, O.M.; Oppenheim, J.J.; Hanisch, F.G.; Finn, O.J. Human tumor antigen MUC1 is chemotactic for immature dendritic cells and elicits maturation but does not promote Th1 type immunity. J. Immunol. 2005, 175, 1628–1635. [Google Scholar] [CrossRef] [PubMed]
  98. Steinman, R.M.; Hawiger, D.; Nussenzweig, M.C. Tolerogenic dendritic cells. Annu. Rev. Immunol. 2003, 21, 685–711. [Google Scholar] [CrossRef] [PubMed]
  99. Ferreira, J.A.; Videira, P.A.; Lima, L.; Pereira, S.; Silva, M.; Carrascal, M.; Severino, P.F.; Fernandes, E.; Almeida, A.; Costa, C.; et al. Overexpression of tumour-associated carbohydrate antigen sialyl-Tn in advanced bladder tumours. Mol. Oncol. 2013, 7, 719–731. [Google Scholar] [CrossRef] [PubMed]
  100. Mantovani, A.; Allavena, P. The interaction of anticancer therapies with tumor-associated macrophages. J. Exp. Med. 2015, 212, 435–445. [Google Scholar] [CrossRef] [PubMed]
  101. Xu, Y.; Zeng, H.; Jin, K.; Liu, Z.; Zhu, Y.; Xu, L.; Wang, Z.; Chang, Y.; Xu, J. Immunosuppressive tumor-associated macrophages expressing interlukin-10 conferred poor prognosis and therapeutic vulnerability in patients with muscle-invasive bladder cancer. J. Immunother. Cancer 2022, 10, e003416. [Google Scholar] [CrossRef]
  102. Hu, W.M.; Li, M.; Ning, J.Z.; Tang, Y.Q.; Song, T.B.; Li, L.Z.; Zou, F.; Cheng, F.; Yu, W.M. FAM171B stabilizes vimentin and enhances CCL2-mediated TAM infiltration to promote bladder cancer progression. J. Exp. Clin. Cancer Res. 2023, 42, 290. [Google Scholar] [CrossRef]
  103. Chen, C.; He, W.; Huang, J.; Wang, B.; Li, H.; Cai, Q.; Su, F.; Bi, J.; Liu, H.; Zhang, B.; et al. LNMAT1 promotes lymphatic metastasis of bladder cancer via CCL2 dependent macrophage recruitment. Nat. Commun. 2018, 9, 3826. [Google Scholar] [CrossRef] [PubMed]
  104. Wu, H.; Zhang, X.; Han, D.; Cao, J.; Tian, J. Tumour-associated macrophages mediate the invasion and metastasis of bladder cancer cells through CXCL8. PeerJ 2020, 8, e8721. [Google Scholar] [CrossRef] [PubMed]
  105. Guo, Q.; Jin, Z.; Yuan, Y.; Liu, R.; Xu, T.; Wei, H.; Xu, X.; He, S.; Chen, S.; Shi, Z.; et al. New Mechanisms of Tumor-Associated Macrophages on Promoting Tumor Progression: Recent Research Advances and Potential Targets for Tumor Immunotherapy. J. Immunol. Res. 2016, 2016, 9720912. [Google Scholar] [CrossRef] [PubMed]
  106. Pignot, G.; Bieche, I.; Vacher, S.; Guet, C.; Vieillefond, A.; Debre, B.; Lidereau, R.; Amsellem-Ouazana, D. Large-scale real-time reverse transcription-PCR approach of angiogenic pathways in human transitional cell carcinoma of the bladder: Identification of VEGFA as a major independent prognostic marker. Eur. Urol. 2009, 56, 678–688. [Google Scholar] [CrossRef]
  107. Shimizu, K.; Nakata, M.; Hirami, Y.; Yukawa, T.; Maeda, A.; Tanemoto, K. Tumor-infiltrating Foxp3+ regulatory T cells are correlated with cyclooxygenase-2 expression and are associated with recurrence in resected non-small cell lung cancer. J. Thorac. Oncol. 2010, 5, 585–590. [Google Scholar] [CrossRef]
  108. Pichler, R.; Fritz, J.; Zavadil, C.; Schafer, G.; Culig, Z.; Brunner, A. Tumor-infiltrating immune cell subpopulations influence the oncologic outcome after intravesical Bacillus Calmette-Guerin therapy in bladder cancer. Oncotarget 2016, 7, 39916–39930. [Google Scholar] [CrossRef]
  109. Masopust, D.; Vezys, V.; Wherry, E.J.; Ahmed, R. A brief history of CD8 T cells. Eur. J. Immunol. 2007, 37 (Suppl. S1), S103–S110. [Google Scholar] [CrossRef]
  110. Mohammed, S.A.; Hetta, H.F.; Zahran, A.M.; Tolba, M.E.M.; Attia, R.A.H.; Behnsawy, H.M.; Algammal, A.M.; Batiha, G.E.; Mohammed, A.Q.; Ahmad, A.A. T cell subsets, regulatory T, regulatory B cells and proinflammatory cytokine profile in Schistosoma haematobium associated bladder cancer: First report from Upper Egypt. PLoS Negl. Trop. Dis. 2023, 17, e0011258. [Google Scholar] [CrossRef]
  111. Quinn, K.M.; Rich, F.J.; Goldsack, L.M.; de Lisle, G.W.; Buddle, B.M.; Delahunt, B.; Kirman, J.R. Accelerating the secondary immune response by inactivating CD4(+)CD25(+) T regulatory cells prior to BCG vaccination does not enhance protection against tuberculosis. Eur. J. Immunol. 2008, 38, 695–705. [Google Scholar] [CrossRef]
  112. Bhattacharya, D.; Dwivedi, V.P.; Kumar, S.; Reddy, M.C.; Van Kaer, L.; Moodley, P.; Das, G. Simultaneous inhibition of T helper 2 and T regulatory cell differentiation by small molecules enhances Bacillus Calmette-Guerin vaccine efficacy against tuberculosis. J. Biol. Chem. 2014, 289, 33404–33411. [Google Scholar] [CrossRef]
  113. Huang, H.; Wang, Z.; Zhang, Y.; Pradhan, R.N.; Ganguly, D.; Chandra, R.; Murimwa, G.; Wright, S.; Gu, X.; Maddipati, R.; et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell 2022, 40, 656–673.e7. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, S.Z.; Roden, D.L.; Wang, C.; Holliday, H.; Harvey, K.; Cazet, A.S.; Murphy, K.J.; Pereira, B.; Al-Eryani, G.; Bartonicek, N.; et al. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. EMBO J. 2020, 39, e104063. [Google Scholar] [CrossRef] [PubMed]
  115. Chakravarthy, A.; Khan, L.; Bensler, N.P.; Bose, P.; De Carvalho, D.D. TGF-beta-associated extracellular matrix genes link cancer-associated fibroblasts to immune evasion and immunotherapy failure. Nat. Commun. 2018, 9, 4692. [Google Scholar] [CrossRef]
  116. Kuen, J.; Darowski, D.; Kluge, T.; Majety, M. Pancreatic cancer cell/fibroblast co-culture induces M2 like macrophages that influence therapeutic response in a 3D model. PLoS ONE 2017, 12, e0182039. [Google Scholar] [CrossRef] [PubMed]
  117. Zhuang, J.; Lu, Q.; Shen, B.; Huang, X.; Shen, L.; Zheng, X.; Huang, R.; Yan, J.; Guo, H. TGFbeta1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci. Rep. 2015, 5, 11924. [Google Scholar] [CrossRef] [PubMed]
  118. Dou, D.; Ren, X.; Han, M.; Xu, X.; Ge, X.; Gu, Y.; Wang, X. Cancer-Associated Fibroblasts-Derived Exosomes Suppress Immune Cell Function in Breast Cancer via the miR-92/PD-L1 Pathway. Front. Immunol. 2020, 11, 2026. [Google Scholar] [CrossRef] [PubMed]
  119. Harper, J.; Sainson, R.C. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin. Cancer Biol. 2014, 25, 69–77. [Google Scholar] [CrossRef]
  120. Navab, R.; Strumpf, D.; Bandarchi, B.; Zhu, C.Q.; Pintilie, M.; Ramnarine, V.R.; Ibrahimov, E.; Radulovich, N.; Leung, L.; Barczyk, M.; et al. Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer. Proc. Natl. Acad. Sci. USA 2011, 108, 7160–7165. [Google Scholar] [CrossRef]
  121. Herrera, M.; Islam, A.B.; Herrera, A.; Martin, P.; Garcia, V.; Silva, J.; Garcia, J.M.; Salas, C.; Casal, I.; de Herreros, A.G.; et al. Functional heterogeneity of cancer-associated fibroblasts from human colon tumors shows specific prognostic gene expression signature. Clin. Cancer Res. 2013, 19, 5914–5926. [Google Scholar] [CrossRef]
  122. Noma, K.; Smalley, K.S.; Lioni, M.; Naomoto, Y.; Tanaka, N.; El-Deiry, W.; King, A.J.; Nakagawa, H.; Herlyn, M. The essential role of fibroblasts in esophageal squamous cell carcinoma-induced angiogenesis. Gastroenterology 2008, 134, 1981–1993. [Google Scholar] [CrossRef]
  123. Grum-Schwensen, B.; Klingelhofer, J.; Berg, C.H.; El-Naaman, C.; Grigorian, M.; Lukanidin, E.; Ambartsumian, N. Suppression of tumor development and metastasis formation in mice lacking the S100A4(mts1) gene. Cancer Res. 2005, 65, 3772–3780. [Google Scholar] [CrossRef]
  124. Gorchs, L.; Fernandez Moro, C.; Bankhead, P.; Kern, K.P.; Sadeak, I.; Meng, Q.; Rangelova, E.; Kaipe, H. Human Pancreatic Carcinoma-Associated Fibroblasts Promote Expression of Co-inhibitory Markers on CD4(+) and CD8(+) T-Cells. Front. Immunol. 2019, 10, 847. [Google Scholar] [CrossRef]
  125. Li, Z.; Zhou, J.; Zhang, J.; Li, S.; Wang, H.; Du, J. Cancer-associated fibroblasts promote PD-L1 expression in mice cancer cells via secreting CXCL5. Int. J. Cancer 2019, 145, 1946–1957. [Google Scholar] [CrossRef]
  126. Nazareth, M.R.; Broderick, L.; Simpson-Abelson, M.R.; Kelleher, R.J., Jr.; Yokota, S.J.; Bankert, R.B. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. J. Immunol. 2007, 178, 5552–5562. [Google Scholar] [CrossRef]
  127. Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8 (+) T Cells to protect tumour cells. Nat. Commun. 2018, 9, 948. [Google Scholar] [CrossRef] [PubMed]
  128. Liang, T.; Tao, T.; Wu, K.; Liu, L.; Xu, W.; Zhou, D.; Fang, H.; Ding, Q.; Huang, G.; Wu, S. Cancer-Associated Fibroblast-Induced Remodeling of Tumor Microenvironment in Recurrent Bladder Cancer. Adv. Sci. 2023, 10, e2303230. [Google Scholar] [CrossRef] [PubMed]
  129. Goulet, C.R.; Champagne, A.; Bernard, G.; Vandal, D.; Chabaud, S.; Pouliot, F.; Bolduc, S. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019, 19, 137. [Google Scholar] [CrossRef]
  130. Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef] [PubMed]
  131. Zhang, Z.; Yu, Y.; Zhang, Z.; Li, D.; Liang, Z.; Wang, L.; Chen, Y.; Liang, Y.; Niu, H. Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1. J. Exp. Clin. Cancer Res. 2023, 42, 316. [Google Scholar] [CrossRef]
  132. Kovacs, S.A.; Fekete, J.T.; Gyorffy, B. Predictive biomarkers of immunotherapy response with pharmacological applications in solid tumors. Acta Pharmacol. Sin. 2023, 44, 1879–1889. [Google Scholar] [CrossRef]
  133. Hannouneh, Z.A.; Hijazi, A.; Alsaleem, A.A.; Hami, S.; Kheyrbek, N.; Tanous, F.; Khaddour, K.; Abbas, A.; Alshehabi, Z. Novel immunotherapeutic options for BCG-unresponsive high-risk non-muscle-invasive bladder cancer. Cancer Med. 2023, 12, 21944–21968. [Google Scholar] [CrossRef] [PubMed]
  134. van Dorp, J.; Pipinikas, C.; Suelmann, B.B.M.; Mehra, N.; van Dijk, N.; Marsico, G.; van Montfoort, M.L.; Hackinger, S.; Braaf, L.M.; Amarante, T.; et al. High- or low-dose preoperative ipilimumab plus nivolumab in stage III urothelial cancer: The phase 1B NABUCCO trial. Nat. Med. 2023, 29, 588–592. [Google Scholar] [CrossRef]
  135. Tu, M.M.; Ng, T.L.; De Jong, F.C.; Zuiverloon, T.C.M.; Fazzari, F.G.T.; Theodorescu, D. Molecular Biomarkers of Response to PD-1/PD-L1 Immune Checkpoint Blockade in Advanced Bladder Cancer. Bladder Cancer 2019, 5, 131–145. [Google Scholar] [CrossRef] [PubMed]
  136. Tan, W.P.; Tan, W.S.; Inman, B.A. PD-L1/PD-1 Biomarker for Metastatic Urothelial Cancer that Progress Post-platinum Therapy: A Systematic Review and Meta-analysis. Bladder Cancer 2019, 5, 211–223. [Google Scholar] [CrossRef] [PubMed]
  137. Lavoie, J.M.; Black, P.C.; Eigl, B.J. Predictive Biomarkers for Checkpoint Blockade in Urothelial Cancer: A Systematic Review. J. Urol. 2019, 202, 49–56. [Google Scholar] [CrossRef]
  138. Ward Grados, D.F.; Ahmadi, H.; Griffith, T.S.; Warlick, C.A. Immunotherapy for Bladder Cancer: Latest Advances and Ongoing Clinical Trials. Immunol. Investig. 2022, 51, 2226–2251. [Google Scholar] [CrossRef] [PubMed]
  139. Roche, H.-L. A Phase Ib/II, Open-Label Study of the Safety and Pharmacology of Atezolizumab Administered with or without Bacille Calmette-Guérin in Patients with High-Risk Non-Muscle-invasive Bladder Cancer. 2021. Available online: https://clinicaltrials.gov/study/NCT02792192 (accessed on 22 August 2023).
  140. Fiegle, E.; Doleschel, D.; Koletnik, S.; Rix, A.; Weiskirchen, R.; Borkham-Kamphorst, E.; Kiessling, F.; Lederle, W. Dual CTLA-4 and PD-L1 Blockade Inhibits Tumor Growth and Liver Metastasis in a Highly Aggressive Orthotopic Mouse Model of Colon Cancer. Neoplasia 2019, 21, 932–944. [Google Scholar] [CrossRef]
  141. Mangsbo, S.M.; Sandin, L.C.; Anger, K.; Korman, A.J.; Loskog, A.; Totterman, T.H. Enhanced tumor eradication by combining CTLA-4 or PD-1 blockade with CpG therapy. J. Immunother. 2010, 33, 225–235. [Google Scholar] [CrossRef]
  142. van Dijk, N.; Gil-Jimenez, A.; Silina, K.; Hendricksen, K.; Smit, L.A.; de Feijter, J.M.; van Montfoort, M.L.; van Rooijen, C.; Peters, D.; Broeks, A.; et al. Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: The NABUCCO trial. Nat. Med. 2020, 26, 1839–1844. [Google Scholar] [CrossRef]
  143. Loustau, M.; Anna, F.; Drean, R.; Lecomte, M.; Langlade-Demoyen, P.; Caumartin, J. HLA-G Neo-Expression on Tumors. Front. Immunol. 2020, 11, 1685. [Google Scholar] [CrossRef]
  144. Carosella, E.D.; Gregori, S.; Tronik-Le Roux, D. HLA-G/LILRBs: A Cancer Immunotherapy Challenge. Trends Cancer 2021, 7, 389–392. [Google Scholar] [CrossRef] [PubMed]
  145. Lin, A.; Chen, H.X.; Zhu, C.C.; Zhang, X.; Xu, H.H.; Zhang, J.G.; Wang, Q.; Zhou, W.J.; Yan, W.H. Aberrant human leucocyte antigen-G expression and its clinical relevance in hepatocellular carcinoma. J. Cell Mol. Med. 2010, 14, 2162–2171. [Google Scholar] [CrossRef]
  146. Pazmany, L.; Mandelboim, O.; Vales-Gomez, M.; Davis, D.M.; Reyburn, H.T.; Strominger, J.L. Protection from natural killer cell-mediated lysis by HLA-G expression on target cells. Science 1996, 274, 792–795. [Google Scholar] [CrossRef] [PubMed]
  147. Wiendl, H.; Mitsdoerffer, M.; Hofmeister, V.; Wischhusen, J.; Bornemann, A.; Meyermann, R.; Weiss, E.H.; Melms, A.; Weller, M. A functional role of HLA-G expression in human gliomas: An alternative strategy of immune escape. J. Immunol. 2002, 168, 4772–4780. [Google Scholar] [CrossRef] [PubMed]
  148. Lin, A.; Yan, W.H.; Xu, H.H.; Gan, M.F.; Cai, J.F.; Zhu, M.; Zhou, M.Y. HLA-G expression in human ovarian carcinoma counteracts NK cell function. Ann. Oncol. 2007, 18, 1804–1809. [Google Scholar] [CrossRef]
  149. Jasinski-Bergner, S.; Eckstein, M.; Taubert, H.; Wach, S.; Fiebig, C.; Strick, R.; Hartmann, A.; Seliger, B. The Human Leukocyte Antigen G as an Immune Escape Mechanism and Novel Therapeutic Target in Urological Tumors. Front. Immunol. 2022, 13, 811200. [Google Scholar] [CrossRef]
  150. Morandi, F.; Airoldi, I. HLA-G and Other Immune Checkpoint Molecules as Targets for Novel Combined Immunotherapies. Int. J. Mol. Sci. 2022, 23, 2925. [Google Scholar] [CrossRef]
  151. Mukherjee, S.; Patra, R.; Behzadi, P.; Masotti, A.; Paolini, A.; Sarshar, M. Toll-like receptor-guided therapeutic intervention of human cancers: Molecular and immunological perspectives. Front. Immunol. 2023, 14, 1244345. [Google Scholar] [CrossRef]
  152. Kashani, B.; Zandi, Z.; Bashash, D.; Zaghal, A.; Momeny, M.; Poursani, E.M.; Pourbagheri-Sigaroodi, A.; Mousavi, S.A.; Ghaffari, S.H. Small molecule inhibitor of TLR4 inhibits ovarian cancer cell proliferation: New insight into the anticancer effect of TAK-242 (Resatorvid). Cancer Chemother. Pharmacol. 2020, 85, 47–59. [Google Scholar] [CrossRef]
  153. Zhou, J.; Liu, Q.; Qian, R.; Liu, S.; Hu, W.; Liu, Z. Paeonol antagonizes oncogenesis of osteosarcoma by inhibiting the function of TLR4/MAPK/NF-kappaB pathway. Acta Histochem. 2020, 122, 151455. [Google Scholar] [CrossRef]
  154. Cen, X.; Zhu, G.; Yang, J.; Yang, J.; Guo, J.; Jin, J.; Nandakumar, K.S.; Yang, W.; Yin, H.; Liu, S.; et al. TLR1/2 Specific Small-Molecule Agonist Suppresses Leukemia Cancer Cell Growth by Stimulating Cytotoxic T Lymphocytes. Adv. Sci. 2019, 6, 1802042. [Google Scholar] [CrossRef]
  155. Yang, Y.; Li, H.; Fotopoulou, C.; Cunnea, P.; Zhao, X. Toll-like receptor-targeted anti-tumor therapies: Advances and challenges. Front. Immunol. 2022, 13, 1049340. [Google Scholar] [CrossRef]
  156. Zandi, Z.; Kashani, B.; Poursani, E.M.; Bashash, D.; Kabuli, M.; Momeny, M.; Mousavi-Pak, S.H.; Sheikhsaran, F.; Alimoghaddam, K.; Mousavi, S.A.; et al. TLR4 blockade using TAK-242 suppresses ovarian and breast cancer cells invasion through the inhibition of extracellular matrix degradation and epithelial-mesenchymal transition. Eur. J. Pharmacol. 2019, 853, 256–263. [Google Scholar] [CrossRef] [PubMed]
  157. So, E.Y.; Ouchi, T. The application of Toll like receptors for cancer therapy. Int. J. Biol. Sci. 2010, 6, 675–681. [Google Scholar] [CrossRef] [PubMed]
  158. Pahlavanneshan, S.; Sayadmanesh, A.; Ebrahimiyan, H.; Basiri, M. Toll-Like Receptor-Based Strategies for Cancer Immunotherapy. J. Immunol. Res. 2021, 2021, 9912188. [Google Scholar] [CrossRef] [PubMed]
  159. Melssen, M.M.; Petroni, G.R.; Chianese-Bullock, K.A.; Wages, N.A.; Grosh, W.W.; Varhegyi, N.; Smolkin, M.E.; Smith, K.T.; Galeassi, N.V.; Deacon, D.H.; et al. A multipeptide vaccine plus toll-like receptor agonists LPS or polyICLC in combination with incomplete Freund’s adjuvant in melanoma patients. J. Immunother. Cancer 2019, 7, 163. [Google Scholar] [CrossRef] [PubMed]
  160. Pavlick, A.; Blazquez, A.B.; Meseck, M.; Lattanzi, M.; Ott, P.A.; Marron, T.U.; Holman, R.M.; Mandeli, J.; Salazar, A.M.; McClain, C.B.; et al. Combined Vaccination with NY-ESO-1 Protein, Poly-ICLC, and Montanide Improves Humoral and Cellular Immune Responses in Patients with High-Risk Melanoma. Cancer Immunol. Res. 2020, 8, 70–80. [Google Scholar] [CrossRef] [PubMed]
  161. Camargo, J.A.; Passos, G.R.; Ferrari, K.L.; Billis, A.; Saad, M.J.A.; Reis, L.O. Intravesical Immunomodulatory Imiquimod Enhances Bacillus Calmette-Guerin Downregulation of Nonmuscle-invasive Bladder Cancer. Clin. Genitourin. Cancer 2018, 16, e587–e593. [Google Scholar] [CrossRef]
  162. Falke, J.; Hulsbergen-van de Kaa, C.A.; Maj, R.; Oosterwijk, E.; Witjes, J.A. A placebo-controlled efficacy study of the intravesical immunomodulators TMX-101 and TMX-202 in an orthotopic bladder cancer rat model. World J. Urol. 2018, 36, 1719–1725. [Google Scholar] [CrossRef]
  163. Mangsbo, S.M.; Ninalga, C.; Essand, M.; Loskog, A.; Totterman, T.H. CpG therapy is superior to BCG in an orthotopic bladder cancer model and generates CD4+ T-cell immunity. J. Immunother. 2008, 31, 34–42. [Google Scholar] [CrossRef]
  164. Codolo, G.; Fassan, M.; Munari, F.; Volpe, A.; Bassi, P.; Rugge, M.; Pagano, F.; D’Elios, M.M.; de Bernard, M. HP-NAP inhibits the growth of bladder cancer in mice by activating a cytotoxic Th1 response. Cancer Immunol. Immunother. 2012, 61, 31–40. [Google Scholar] [CrossRef] [PubMed]
  165. Singh, V.; Srivastava, N.; Kapoor, R.; Mittal, R.D. Single-nucleotide polymorphisms in genes encoding toll-like receptor -2, -3, -4, and -9 in a case-control study with bladder cancer susceptibility in a North Indian population. Arch. Med. Res. 2013, 44, 54–61. [Google Scholar] [CrossRef] [PubMed]
  166. Cascone, T.; McKenzie, J.A.; Mbofung, R.M.; Punt, S.; Wang, Z.; Xu, C.; Williams, L.J.; Wang, Z.; Bristow, C.A.; Carugo, A.; et al. Increased Tumor Glycolysis Characterizes Immune Resistance to Adoptive T Cell Therapy. Cell Metab. 2018, 27, 977–987.e4. [Google Scholar] [CrossRef] [PubMed]
  167. Kurtova, A.V.; Xiao, J.; Mo, Q.; Pazhanisamy, S.; Krasnow, R.; Lerner, S.P.; Chen, F.; Roh, T.T.; Lay, E.; Ho, P.L.; et al. Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance. Nature 2015, 517, 209–213. [Google Scholar] [CrossRef] [PubMed]
  168. Shirahama, T.; Sakakura, C. Overexpression of cyclooxygenase-2 in squamous cell carcinoma of the urinary bladder. Clin. Cancer Res. 2001, 7, 558–561. [Google Scholar] [PubMed]
  169. Sharma, S.; Yang, S.C.; Zhu, L.; Reckamp, K.; Gardner, B.; Baratelli, F.; Huang, M.; Batra, R.K.; Dubinett, S.M. Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4+ CD25+ T regulatory cell activities in lung cancer. Cancer Res. 2005, 65, 5211–5220. [Google Scholar] [CrossRef]
  170. Dowd, N.P.; Scully, M.; Adderley, S.R.; Cunningham, A.J.; Fitzgerald, D.J. Inhibition of cyclooxygenase-2 aggravates doxorubicin-mediated cardiac injury in vivo. J. Clin. Investig. 2001, 108, 585–590. [Google Scholar] [CrossRef]
  171. Levesque, L.E.; Brophy, J.M.; Zhang, B. Time variations in the risk of myocardial infarction among elderly users of COX-2 inhibitors. CMAJ 2006, 174, 1563–1569. [Google Scholar] [CrossRef]
  172. Cao, Y.; Stosiek, P.; Springer, G.F.; Karsten, U. Thomsen-Friedenreich-related carbohydrate antigens in normal adult human tissues: A systematic and comparative study. Histochem. Cell Biol. 1996, 106, 197–207. [Google Scholar] [CrossRef]
  173. Palucka, K.; Banchereau, J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013, 39, 38–48. [Google Scholar] [CrossRef] [PubMed]
  174. Lima, L.; Neves, M.; Oliveira, M.I.; Dieguez, L.; Freitas, R.; Azevedo, R.; Gaiteiro, C.; Soares, J.; Ferreira, D.; Peixoto, A.; et al. Sialyl-Tn identifies muscle-invasive bladder cancer basal and luminal subtypes facing decreased survival, being expressed by circulating tumor cells and metastases. Urol. Oncol. 2017, 35, 675.e1–675.e8. [Google Scholar] [CrossRef] [PubMed]
  175. Lazennec, G.; Richmond, A. Chemokines and chemokine receptors: New insights into cancer-related inflammation. Trends Mol. Med. 2010, 16, 133–144. [Google Scholar] [CrossRef] [PubMed]
  176. Vindrieux, D.; Escobar, P.; Lazennec, G. Emerging roles of chemokines in prostate cancer. Endocr. Relat. Cancer 2009, 16, 663–673. [Google Scholar] [CrossRef] [PubMed]
  177. Ben-Baruch, A. Organ selectivity in metastasis: Regulation by chemokines and their receptors. Clin. Exp. Metastasis 2008, 25, 345–356. [Google Scholar] [CrossRef] [PubMed]
  178. Nazari, A.; Khorramdelazad, H.; Hassanshahi, G. Biological/pathological functions of the CXCL12/CXCR4/CXCR7 axes in the pathogenesis of bladder cancer. Int. J. Clin. Oncol. 2017, 22, 991–1000. [Google Scholar] [CrossRef] [PubMed]
  179. Ene-Obong, A.; Clear, A.J.; Watt, J.; Wang, J.; Fatah, R.; Riches, J.C.; Marshall, J.F.; Chin-Aleong, J.; Chelala, C.; Gribben, J.G.; et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013, 145, 1121–1132. [Google Scholar] [CrossRef] [PubMed]
  180. Yang, H.; Zhang, Q.; Xu, M.; Wang, L.; Chen, X.; Feng, Y.; Li, Y.; Zhang, X.; Cui, W.; Jia, X. CCL2-CCR2 axis recruits tumor associated macrophages to induce immune evasion through PD-1 signaling in esophageal carcinogenesis. Mol. Cancer 2020, 19, 41. [Google Scholar] [CrossRef] [PubMed]
  181. Huang, Y.H.; Cai, K.; Xu, P.P.; Wang, L.; Huang, C.X.; Fang, Y.; Cheng, S.; Sun, X.J.; Liu, F.; Huang, J.Y.; et al. CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct. Target. Ther. 2021, 6, 10. [Google Scholar] [CrossRef]
  182. Ha, H.; Debnath, B.; Neamati, N. Role of the CXCL8-CXCR1/2 Axis in Cancer and Inflammatory Diseases. Theranostics 2017, 7, 1543–1588. [Google Scholar] [CrossRef]
  183. Fang, W.; Ye, L.; Shen, L.; Cai, J.; Huang, F.; Wei, Q.; Fei, X.; Chen, X.; Guan, H.; Wang, W.; et al. Tumor-associated macrophages promote the metastatic potential of thyroid papillary cancer by releasing CXCL8. Carcinogenesis 2014, 35, 1780–1787. [Google Scholar] [CrossRef]
  184. Huang, Z.; Zhang, M.; Chen, G.; Wang, W.; Zhang, P.; Yue, Y.; Guan, Z.; Wang, X.; Fan, J. Bladder cancer cells interact with vascular endothelial cells triggering EGFR signals to promote tumor progression. Int. J. Oncol. 2019, 54, 1555–1566. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bladder cancer evades immune responses by forming an immunosuppressive microenvironment. (A). PD-1 regulates T cell effector functions and acts as a brake on T cell immune responses. HLA-G-induced inhibitory cells have long-term immune regulatory functions, inducing the generation of suppressive cells and blocking immune effector mechanisms. (B). IL-10-positive TAMs showed an immunosuppressive phenotype resembling that of M2 macrophages within the context of MIBC. STn interferes with the expression of MHC-Ⅱ and co-stimulatory molecules in DCs, reducing their ability to display cancer-associated antigens to T cells. (C). PGE2 interferes with anti-tumor immune responses by affecting the function of immune cells such as T cells, DCs, macrophages, and NK cells. (D). Lactate contributes to creating an immunosuppressive TME by reducing the presence of NK cells and cytotoxic T cells, while suppressing the expression of IFN-γ. PD-1, programmed cell death protein 1; HLA-G, human leukocyte antigen G; TAMs, tumor-associated macrophages; MIBC, muscle-invasive bladder cancer; STn, Sialyl Tn; PGE2, prostaglandin E2; DCs, dendritic cells; NK cells, natural killer cells; IFN-γ, interferon- γ.
Figure 1. Bladder cancer evades immune responses by forming an immunosuppressive microenvironment. (A). PD-1 regulates T cell effector functions and acts as a brake on T cell immune responses. HLA-G-induced inhibitory cells have long-term immune regulatory functions, inducing the generation of suppressive cells and blocking immune effector mechanisms. (B). IL-10-positive TAMs showed an immunosuppressive phenotype resembling that of M2 macrophages within the context of MIBC. STn interferes with the expression of MHC-Ⅱ and co-stimulatory molecules in DCs, reducing their ability to display cancer-associated antigens to T cells. (C). PGE2 interferes with anti-tumor immune responses by affecting the function of immune cells such as T cells, DCs, macrophages, and NK cells. (D). Lactate contributes to creating an immunosuppressive TME by reducing the presence of NK cells and cytotoxic T cells, while suppressing the expression of IFN-γ. PD-1, programmed cell death protein 1; HLA-G, human leukocyte antigen G; TAMs, tumor-associated macrophages; MIBC, muscle-invasive bladder cancer; STn, Sialyl Tn; PGE2, prostaglandin E2; DCs, dendritic cells; NK cells, natural killer cells; IFN-γ, interferon- γ.
Ijms 25 03105 g001
Figure 2. Potential therapeutic strategies for modulating immune evasion. This diagram summarizes candidate factors for treatment using the various mechanisms that bladder cancer uses to evade immune responses. (Red numbers indicate the sequence of results when the target is blocked, and an x means that the path is suppressed.) Targeting the STn can enhance the expression of MHC class II and costimulatory molecules in DCs and their ability to present antigens to T cells. PD-L1/PD-1 inhibitors can increase the levels of CD4+ and CD8+ T cells, reduce Tregs, and induce macrophage M1 polarization. Targeting HLA-G can upregulate PD-1 expression on T lymphocytes, block angiogenesis, and activate NK cells. Targeting mPGES1 and employing targeted genetic enhancement of 15-PGDH can reduce PD-L1-mediated immunosuppression. Blocking LDH-A can increase the production of IFN-γ and granzyme B in NK cells and CD8+ T cells and enhance anti-tumor immune responses to immune checkpoint inhibitors. Inhibiting CXCL12 produced by CAFs can significantly reduce the effect of CAFs on PD-L1 expression and promote the migration of CD8+ T cells toward the tumor stromal area. Targeting the FAM171B protein, which can upregulate CCL2 expression in bladder cancer, can suppress M2 polarization of macrophages and Tregs. STn, Sialyl Tn; DCs, dendritic cells; PD-L1, programmed cell death-ligand 1; PD-1, programmed cell death protein 1; Tregs, regulatory T cells; HLA-G, human leukocyte antigen G; NK cells, natural killer cells; mPGES1, PGE2-producing enzyme microsomal prostaglandin E synthase-1; 15-PGDH, PGE2-degrading enzyme NAD+ dependent 15-hydroxyprostaglandin dehydrogenase; LDH-A, lactate dehydrogenase-A; IFN-γ, interferon-γ; CXCL12, C-X-C motif chemokine 12; CAFs, cancer-associated fibroblasts; CCL2, chemokine(C-C motif) ligand 2; Tregs, regulatory T cells.
Figure 2. Potential therapeutic strategies for modulating immune evasion. This diagram summarizes candidate factors for treatment using the various mechanisms that bladder cancer uses to evade immune responses. (Red numbers indicate the sequence of results when the target is blocked, and an x means that the path is suppressed.) Targeting the STn can enhance the expression of MHC class II and costimulatory molecules in DCs and their ability to present antigens to T cells. PD-L1/PD-1 inhibitors can increase the levels of CD4+ and CD8+ T cells, reduce Tregs, and induce macrophage M1 polarization. Targeting HLA-G can upregulate PD-1 expression on T lymphocytes, block angiogenesis, and activate NK cells. Targeting mPGES1 and employing targeted genetic enhancement of 15-PGDH can reduce PD-L1-mediated immunosuppression. Blocking LDH-A can increase the production of IFN-γ and granzyme B in NK cells and CD8+ T cells and enhance anti-tumor immune responses to immune checkpoint inhibitors. Inhibiting CXCL12 produced by CAFs can significantly reduce the effect of CAFs on PD-L1 expression and promote the migration of CD8+ T cells toward the tumor stromal area. Targeting the FAM171B protein, which can upregulate CCL2 expression in bladder cancer, can suppress M2 polarization of macrophages and Tregs. STn, Sialyl Tn; DCs, dendritic cells; PD-L1, programmed cell death-ligand 1; PD-1, programmed cell death protein 1; Tregs, regulatory T cells; HLA-G, human leukocyte antigen G; NK cells, natural killer cells; mPGES1, PGE2-producing enzyme microsomal prostaglandin E synthase-1; 15-PGDH, PGE2-degrading enzyme NAD+ dependent 15-hydroxyprostaglandin dehydrogenase; LDH-A, lactate dehydrogenase-A; IFN-γ, interferon-γ; CXCL12, C-X-C motif chemokine 12; CAFs, cancer-associated fibroblasts; CCL2, chemokine(C-C motif) ligand 2; Tregs, regulatory T cells.
Ijms 25 03105 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shin, J.; Park, J.W.; Kim, S.Y.; Lee, J.H.; Choi, W.S.; Kim, H.S. Strategies for Overcoming Immune Evasion in Bladder Cancer. Int. J. Mol. Sci. 2024, 25, 3105. https://doi.org/10.3390/ijms25063105

AMA Style

Shin J, Park JW, Kim SY, Lee JH, Choi WS, Kim HS. Strategies for Overcoming Immune Evasion in Bladder Cancer. International Journal of Molecular Sciences. 2024; 25(6):3105. https://doi.org/10.3390/ijms25063105

Chicago/Turabian Style

Shin, Juhyun, Jeong Won Park, Seon Young Kim, Jun Ho Lee, Wahn Soo Choi, and Hyuk Soon Kim. 2024. "Strategies for Overcoming Immune Evasion in Bladder Cancer" International Journal of Molecular Sciences 25, no. 6: 3105. https://doi.org/10.3390/ijms25063105

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop