1. Introduction
Bladder cancer (BC) accounts for approximately 3% of global cancer diagnoses. It was recently the 12th most commonly diagnosed cancer and the 14th leading cause of cancer-related death worldwide [
1]. Approximately 25% of BC cases are muscle-invasive BC (MIBC) cases [
2]. The current therapy for MIBC consists of systemic chemotherapy and/or immunotherapy, radical treatment (cystectomy or radiotherapy), or palliation [
3,
4]. In addition, trimodality therapy, i.e., maximal endoscopic transurethral resection of the bladder tumor followed by concurrent chemo-radiotherapy as an alternative to radical cystectomy, has been discussed [
5]. The degree of lymph node involvement and tumor stage are prognostic factors for MIBC [
3,
6]. Adjuvant chemotherapy was effective in lymph node-positive MIBC patients regardless of their p53 status [
7]. Protein and glycoprotein biomarkers are a demonstrably viable option in BC diagnostics [
8]. However, there are still no applied prognostic and/or predictive protein biomarkers for chemotherapy response in MIBC patients.
Many studies, including our own, have reported that the tumor immune microenvironment is also associated with survival [
5,
9,
10,
11,
12]. In particular, the presence of tumor-infiltrating immune cells, including lymphocytes identified by their protein or gene expression profile, is associated with superior 5-year overall survival (OS) or disease-specific survival (DSS) [
9,
10,
12]. However, immune cells can also express immune checkpoint receptors, such as programmed death 1 (PD-1), which play a role in restraining immune system hyperactivation. Cancer cells can hijack this coinhibitory pathway and escape immune surveillance [
13]. Most recently, combination therapy with an antibody–drug conjugate (enfortumab vedotin) directed against a cell surface receptor (nectin-4) and an inhibitor (pembrolizumab) of the immune checkpoint receptor PD-1 resulted in significantly better outcomes than chemotherapy in patients with untreated locally advanced or metastatic urothelial bladder carcinoma [
14]. These findings further support the striking role of the immune microenvironment in the prognosis and therapeutic response of MIBC patients.
Chemokines play a major role in the interaction between cancer cells and the immune microenvironment [
15,
16]. Chemokines have complex functions both in anti-tumor and pro-tumor immune responses, as reviewed in [
17]. Recently, we showed that the protein expression of the chemokine CCL2 (monocyte chemotactic protein 1/MCP-1) in tumor cells (TCs) was an independent negative prognostic factor for overall survival (OS), but its expression in immune cells (ICs) was an independent positive prognostic factor for disease-specific survival (DSS) in MIBC patients [
18]. A major tumor-promoting role for the coexpression of the chemokines CCL2 and CCL5 in tumor cells has been suggested in breast malignancies [
19]. Furthermore, in breast cancer, CCL2 and CCL5 expression is restricted not only to tumor cells but also to cells of the tumor microenvironment, including fibroblasts, endothelial cells, mesenchymal stem cells, smooth muscle cells and immune cells such as tumor-associated macrophages and T cells [
20].
The CCL5 gene was first described by Schall et al. [
21]. It encodes a T-cell-specific molecule that Schall et al. termed RANTES (an acronym for regulated upon activation, normally T-expressed, and presumably secreted). It belongs to the CC (cysteine–cysteine) motif subfamily of chemokines and is involved in intercellular communication [
22,
23]. CCL5 is overexpressed in many tumor types, such as breast cancer, pancreatic cancer, colorectal carcinoma, esophageal cancer, prostate cancer, lung cancer, gastric adenocarcinoma, melanoma, head and neck cancer, acute lymphocytic leukemia, Hodgkin lymphoma, multiple myeloma, chondrosarcoma, and osteosarcoma, as reviewed in [
24]. Downstream pathways of CCL5 and its main receptor CCR5 include the PI3K/AKT, NF-kB, HIF-a, RAS-ERK-MEK, JAK-STAT and TGF-β-Smad pathways, which are associated with cell proliferation, angiogenesis, apoptosis, invasion, metastasis, and inflammation, as reviewed in [
25].
In this study, we investigated whether the chemokine CCL5 is associated with prognosis when expressed in TCs or ICs in MIBC patients and whether this association is comparable to that of the chemokine CCL2. In addition, we were interested in whether CCL5 can be used as a predictive marker for chemotherapy response.
3. Discussion
In our study, we analyzed the protein expression of CCL5 in TCs and ICs in MIBC patients (n = 175) and assessed its association with clinicopathological and survival data for the first time. Interestingly, CCL5 staining in TCs was weakly positively correlated with CCL5 staining in ICs. Remarkably, CCL5 staining in TCs was positively correlated with the lymph node stage and negatively correlated with survival time and time to recurrence. CCL2 expression in TCs was also correlated with time to recurrence [
18]. In contrast, CCL5 staining in ICs was positively correlated with survival time, time to recurrence, DSS, and RFS. As expected, we observed a strong correlation between immune cell markers and CCL5 expression in ICs. The highest correlations were detected with markers for NK cells and cytotoxic CD8+ cells, which is in agreement with the positive correlation with survival time and time to recurrence in this study and our previous study, where the cytotoxic T-cell-related gene expression signature predicted improved survival in MIBC patients [
9]. In addition, we reported comparable correlations of CCL2 expression in ICs with DSS and RFS [
18]. This concordance suggests a somewhat coordinated coexpression of the chemokines CCL2 and CCL5 in MIBC, as has been reported in breast cancer [
20].
Next, we studied whether CCL5 expression in TCs or in ICs was an independent prognostic marker in all MIBC patients or in subgroups of MIBC patients stratified according to tumor stage, lymph node stage, chemotherapy treatment or molecular subtype.
Multivariate Cox regression analysis showed that CCL5 positivity in TCs was an independent negative prognostic marker for DSS in all MIBC patients. In addition, CCL5 positivity in TCs was an independent prognostic factor for OS, DSS and RFS in MIBC patient subgroups, i.e., in the luminal molecular subtype subgroup. In addition, DSS in the tumor stage 2 and nodal stage N0 subgroups was analyzed. In our previous study of CCL2 expression in the TC of MIBC patients, we found that CCL2 positivity in TCs of the luminal molecular subtype was associated with shorter OS [
18]. In contrast to the previous study, in all patients and in tumor stage 2 patients, CCL2 positivity in TC was associated with shorter OS, while in the chemotherapy-treated patients, CCL2 positivity was associated with shorter OS and DSS. These findings suggest that, although CCL5 and CCL2 positivity in TCs is a negative prognostic factor, this finding can be applied to different MIBC patient groups and prognostic outcomes.
In contrast to the findings in TCs, CCL5 positivity in ICs was an independent positive prognostic parameter for OS, DSS, and RFS in all MIBC patients. In addition, CCL5 positivity was also an independent prognostic parameter in the MIBC subgroups, i.e., for OS, DSS, and RFS in both the tumor stage 3 + 4 and no chemotherapy subgroups. In our previous study of CCL2 expression in ICs, we found that CCL2 IC positivity was associated with a longer DSS for all MIBC patients and for those from the subgroup without chemotherapy. The latter subgroup also exhibited CCL2 positivity in ICs and a longer RFS. However, again somewhat different in the present study, CCL5 positivity in ICs was associated with longer OS, DSS, and RFS, which was not found in a previous study [
18]. This finding suggested that although CCL5 and CCL2 positivity in ICs is a positive prognostic factor, this finding can be applied to different MIBC patient groups and prognostic outcomes.
There are different prognostic impacts of CCL5 staining in TCs and ICs. The CCL5 axis and its main receptor CCR5 support tumor progression through multiple mechanisms, such as increasing tumor growth, inducing extracellular matrix remodeling, enhancing tumor cell migration (metastasis formation), expanding cancer cell stemness, promoting cancer cell resistance to drugs, decreasing cytotoxicity to DNA-damaging agents, deregulating cellular energetics (metabolic reprogramming), and promoting angiogenesis, as reviewed in [
24]. Breast cancer cells can stimulate the de novo secretion of CCL5 from mesenchymal stem cells within the tumor stroma, which then acts in a paracrine fashion on cancer cells to enhance their motility, invasion, and metastasis [
26]. The oncogene MYC, which functions as a transcription factor in many human tumors, elicits the production of chemokines, including CCL2 and CCL5. This attracts inflammatory cells (e.g., mast cells), which promote angiogenesis and tumor growth [
27]. NF-κB activity in breast cancer mouse cells can induce the expression of CCL5, which drives the recruitment of CCR5-expressing macrophages, which supplies breast tumor cells with collagen that promotes their proliferation [
28]. Therapy-induced changes in the expression of chemokines can contribute to tumor resistance or tumor recurrence. The upregulation of CCL2 and CCL5 postradiotherapy results in the recruitment of immunosuppressive cells, such as CCR2 + CCR5+ monocytes, MDSCs, and CCR2+ Treg cells, leading to cancer outgrowth, as reviewed in [
17].
On the other hand, CCL5 is a natural adjuvant for enhancing anti-tumor immune responses [
29]. CCL5 promotes anti-tumor immunity by recruiting anti-tumor T cells and dendritic cells to the tumor microenvironment, and, in this way, it increases the immunotherapy response in different tumor types, as reviewed in [
24]. Together with IL-2 and IFN-γ, which are released by T cells, CCL5 induces the activation and proliferation of particular NK cells to generate C-C chemokine-activated killer cells [
20,
30]. Conversely, stimulated NK cells can produce T-cell-recruiting chemokines, including CCL2 and CCL5, in breast cancer patients [
31].
Our interesting finding of a response to chemotherapy in CCL5-negative IC patients led us to study the chemotherapy response—analyzed by OS, DSS, and RFS—separately in patients with CCL5-positive or CCL5-negative ICs in all subgroups of MIBC patients and the complete MIBC patient cohort. Chemotherapy treatment in all patients, but only in those with CCL5-negative ICs, was an independent positive predictor of OS, DSS and RFS compared to patients who did not receive chemotherapy. In the MIBC subgroup analysis this prognostic effect was also found for OS, DSS, and RFS in CCL5 IC-negative patients in the tumor stage 3 + 4 subgroup or in the luminal molecular subtype subgroup. Furthermore, in IC CCL5-negative patients in the nodal stage N0 subgroup, chemotherapy treatment was associated with a reduced risk of death. However, in CCL5-positive patients in the nodal stage N1/N2 subgroup, chemotherapy treatment was associated with a reduced risk of death compared to patients who did not receive chemotherapy. We suggest that CCL5 could mark a population of ICs that may be anti-tumorigenic in N0 patients but pro-tumorigenic in N1 + 2 MIBC patients. Generally, a tumor, such as bladder cancer, is a key immunological player that can shape immune responses to favor itself [
32]. For an overview of tumor-infiltrating immune cells and their therapeutic implications, we would like to refer to excellent reviews [
17,
33]. A Swedish study of MIBC reported that patients treated with radio-/chemotherapy or radiotherapy had better OS and DSS than untreated patients [
34]; however, in that study, no further stratification (e.g., for immunological markers) was performed.
Overall, chemotherapy was associated with a better prognosis, mostly in CCL5-negative patients. However, chemotherapy was not advantageous for OS in all CCL5 IC-positive patients but was beneficial for OS in the N1/N2 subgroup. In our previous study on the effect of chemotherapy on CCL2 staining in ICs, we found that, in the subgroup with the most aggressive tumors (N1 + 2 and tumor stage 3+ 4), patients with CCL2-positive ICs showed a better response to chemotherapy treatment in terms of OS, DSS, and RFS than MIBC patients indicated with negative IC CCL2 staining [
35]. However, in our previous CCL2 study, chemotherapy was associated with a poorer prognosis (shorter RFS) in patients with CCL2-positive ICs in the N0 subgroup and with poorer survival (shorter OS, DSS, RFS) in patients with CCL2-positive ICs in the pT2 subgroup than in patients who did not receive chemotherapy. Such a significant association between CCL5 IC positivity and poor survival after chemotherapy could be observed only as a trend for nodal stage N0 patients.
We previously showed that a cytotoxic T-cell-related gene expression signature predicts improved survival in MIBC patients after radical cystectomy and adjuvant chemotherapy [
9]. In addition, patients with T-cell-inflamed tumors that are enriched in T-cell-recruiting chemokines, such as CCL5, CXCL9, CXCL10, and CXCL11, are most likely to benefit from checkpoint blockade therapy [
17,
36].
Our finding that MIBC patients with CCL5-positive ICs have a better prognosis (OS, DSS and RFS) suggests that CCL5 expression in ICs, possibly as a surrogate marker for cytotoxic CD8+ T cells and NK cells, may play a role in the anti-tumor immune response. CD8+ T cells can secrete inflammatory chemokines, such as CCL3 and CCL5, which increase infiltration of neutrophils, monocytes, and Th1 lymphocytes. In this way, they can contribute to a so-called auto-recruitment of cytotoxic T cells [
37]. Recently, Sun et al. indicated that CD8+ T cells infiltration can be regulated by a circular RNA (circMGA) that stabilizes CCL5 mRNA in bladder cancer [
38]. Interestingly, treatment with circMGA and anti-PD-1 can synergistically suppress xenograft bladder cancer growth [
38].
There are different hurdles with CCL5/CCR5 inhibition in tumor therapy since CCL5 acts as a double-edged sword—initially fueling tumor development but also recruiting antitumor cell populations to the tumor over time [
39]. For a recent overview of the CCL5/CCR5 network and their clinical application as drug targets, especially in colorectal cancer, we would like to refer to a review by Zhang et al. [
40]. In addition, recent findings by Jacobs et al. are of interest. The suppression of CCL5 expression by heat shock Factor 1 (HSF1) prevents CD8+ T-cell influx, which supports immune-mediated tumor killing [
41]. The authors suggest that targeting HSF1 could improve immunotherapies. Furthermore, the T-cell–inflamed gene expression profile, which includes CCL5, appears to be an emerging predictive biomarker for the pembrolizumab response [
36].
Our study has several limitations. It was a retrospective study, and for a comprehensive statistical analysis of two parameters in eight subgroups, the number of study patients was rather low. In addition, only 24.6% (43/175) of our patients were treated with chemotherapy, which is again a rather small cohort. Ultimately, our results must be evaluated in a larger prospective study. However, altogether, the number of study patients (n = 175) allowed for reasonable multivariate analysis of one parameter, such as CCL5 staining, for its prognostic and predictive relevance in MIBC patients.
Overall, CCL5 positivity in TCs is an independent negative prognostic factor for DSS. In contrast, CCL5 positivity in ICs was significantly associated with improved OS, DSS, and RFS. Chemotherapy treatment was associated with a better prognosis for OS, mostly in CCL5 IC-negative patients, but not in CCL5 IC-positive patients in the N1/N2 subgroup (n = 38). We suggest that CCL5 staining in TCs and ICs seems to be a prognostic marker; additionally, CCL5 detection in ICs might serve as a predictive marker for adjuvant chemotherapy and, possibly, for future immune checkpoint therapy.