Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 6.
Published in final edited form as: Adv Exp Med Biol. 2020;1296:275–290. doi: 10.1007/978-3-030-59038-3_17

The Tumor Microenvironment of Bladder Cancer

Ken Hatogai 1,2,3, Randy F Sweis 1,2,3
PMCID: PMC8345230  NIHMSID: NIHMS1729365  PMID: 34185299

Abstract

Bladder cancer has been well known as immunotherapy-responsive disease as intravesical therapy with BCG has been the standard of care for non-muscle invasive disease for several decades. In addition, immune checkpoint inhibitors have dramatically changed the treatment of metastatic bladder cancer. However, only a small fraction of patients with bladder cancer can benefit from these therapies. As immunotherapies act on the tumor microenvironment, understanding it is essential to expand the efficacy of modern treatments. The bladder cancer microenvironment consists of various components including tumor cells, immune cells, and other stromal cells, affecting each other via immune checkpoint molecules, cytokines, and chemokines. The development of an antitumor immune response depends on tumor antigen recognition by antigen presenting cells and priming and recruitment of effector T cells. Accumulated evidence shows that these processes are impacted by multiple types of immune cells in the tumor microenvironment including regulatory T cells, tumor-associated macrophages, and myeloid derived suppressor cells. In addition, recent advances in genomic profiling have shed light on the relationship between molecular subtypes and the tumor microenvironment. Finally, emerging evidence has shown that multiple factors can impact the tumor microenvironment in bladder cancer, including tumor-oncogenic signaling, patient genetics, and the commensal microbiome.

Keywords: Bladder cancer, Tumor microenvironment, T cell-inflamed, Tumor infiltrating lymphocytes, Urothelial cancer, Macrophages, Myeloid-derived suppressor cells, Dendritic cells, Pericytes, Immune checkpoint inhibitors

17.1. Introduction

Bladder cancer is the most common malignancy of the urinary system, and estimated to cause 165,084 annual deaths worldwide, including 17,240 in the United States [1, 2]. Urothelial carcinoma is the most predominant histological type and accounts for 90% of bladder cancers in the United States and Europe. Recent comprehensive gene expression analyses have identified molecular classifications that further characterize urothelial carcinoma more with more precision. These classifications may have important implications for prognosis and treatment response [37]. Bladder cancer can generally be staged into three categories in accordance with progression, namely non-muscle invasive bladder cancer (NMIBC), muscle invasive bladder cancer (MIBC), and metastatic bladder cancer in relation to treatment. Historically, NMIBC has been well known to be an immunotherapy-responsive cancer type owing to the success of intravesical therapy with Bacillus Calmette-Guerin (BCG). This treatment following transurethral resection of bladder tumors (TURBT) has been the standard therapy for several decades [811]. Despite incomplete understanding of the mechanism of its therapeutic effect, available evidence suggests that the cellular inflammatory response includes immune cells such as CD4+ and CD8+ lymphocytes, natural killer cells, granulocytes, and macrophage as well as cytokines such as interleukin-2 (IL-2), IL-8, IL-12, interferon-gamma (IFNg), tumor necrosis factor (TNF)-alpha, and tumor necrosis factor apoptosis inducing ligand (TRAIL) [1215].

More recently, monoclonal antibodies targeting the PD-1/L1 immune checkpoint pathway were found to be effective for advanced bladder cancer. Since 2016, five immune checkpoint inhibitors targeting this pathway received regulatory approval. However, response rates of these drugs remain less than 25% in the first line setting, and those in the second line setting are only 15–20% without biomarker selection [1622]. Thus, the majority of patients with bladder cancer still fail to respond to immunotherapy. Even in patients with PD-L1 expression by immunohistochemistry, which is currently the most used biomarker for predicting treatment response, the response rates are still less than 50% [21, 22]. Importantly, only a limited proportion of patients have tumors with PD-L1 expression, so the benefit of immune checkpoint inhibitors in bladder cancer has been limited so far.

Immune checkpoint blockade, including those targeting the PD-1/L1 immune checkpoint pathway, is based on the inhibition of the tumor-mediated suppression of antitumor immune response and the promotion of cytotoxic effects of T cells on tumor cells. The presence of an existing antitumor T cell response is necessary for the activity of immunotherapy [23]. Although cytotoxic T cells play a major role in antitumor immune response, their activation and ability to attack tumor cells is regulated by the balance between the stimulatory and inhibitory signals among various types of immune cells and tumor cells in the tumor microenvironment (TME) [24, 25]. Therefore, gaining a deeper understanding of the TME is crucial for advancing the efficacy of immunotherapy. In this review, we summarize the current knowledge about TME focusing on bladder cancer in relation to its component immune cells, including both preclinical clinical data. In addition, we describe recently emerging evidence linking to molecular pathways to the TME.

17.2. Main Text

17.2.1. T Cell-Inflamed and Non-T Cell-Inflamed Tumor Microenvironment

T cell-inflamed tumors are those with a pre-existing antitumor immune response, characterized by cytotoxic T cells and other important immune cells, such as antigen-presenting cells. The presence of negative immune regulators is also typically also be found in T cell-inflamed tumors (Fig. 17.1). These immune microenvironment phenotypes can be identified by gene expression profiling, immunohistochemistry, or other emerging technologies. Divergent phenotypes are characterized by the presence or absence of a gene expression profile indicating presence of an existing antitumor T cell response. These signatures include T cell markers and chemokines necessary for recruiting effector T cells to TME, such as CD8A, CCL4, CXCL9, CXCL10, ICOS, and GZMK [23, 26]. The T cell-inflamed phenotype shows spontaneous T cell priming and recruitment effector T cells in to TME even prior to any immunotherapy treatment. In contrast, non-T cell-inflamed phenotype lacks such T cell response at baseline. Clinically, these phenotypes have been correlated with prognosis and response to immunotherapies such as checkpoint inhibitors [2628]. We previously reported that T cell-inflamed and non-T cell-inflamed phenotypes can be also identified in bladder cancers based on gene expressions of a cluster of 725 genes derived from a T cell-inflamed signature using RNA-Seq gene expression data of the Cancer Genome Atlas (TCGA). These phenotypes were highly correlated with CD8+ T cell infiltration in immunohistochemistry as well as mRNA expression of negative regulatory molecules such as PD-L1, IDO, TIM3, and LAG3 [29]. More in depth immune phenotyping of the TME in bladder cancer is needed, and current research efforts have been focused on this goal.

Fig. 17.1.

Fig. 17.1

T cell-inflamed versus non-T cell-inflamed tumor microenvironments in bladder cancer. T cell-inflamed tumors show infiltration of T cells, antigen presenting cells (APCs), and other immune cells. T cell-inflamed tumors are also characterized by the presence of negative regulators such as PD-L1 expression and regulatory T cell (Treg) infiltration (Treg). Non-T cell inflamed tumors lack infiltrating T cells and immune cells and immune exclusion in the tumor microenvironment may be a result of multiple factors including tumor-intrinsic oncogenic signaling

17.2.2. Cytotoxic T Cells

As cytotoxic CD8+ T cells have an ability to directly attack cancer cells, they have been studied in the bladder cancer TME for more than a decade. Prior reports have concluded that greater infiltration of CD8+ T cells was related to favorable outcomes, such as overall survival, disease-free survival, and disease-specific survival both in patients with MIBC and NMIBC [3032]. The backbone of immunotherapy for advanced bladder cancer is anti-PD-1/PD-L1 antibodies, which enhance cytotoxic activity of CD8+ cytotoxic T cells via blocking the suppressive signal. Thus, expression of PD-1, the receptor for PD-L1 and PD-L2, on T cells is also of interest in the TME. Expression of PD-1 is a marker of T cell exhaustion in the context of chronic antigen stimulation. Infiltration of CD8+ T cells and PD-1+ T cells has been highly correlated in the stromal area, suggesting that an exhausted immune state occurs in TME and is a target of immune checkpoint blockade [33, 34]. According to recently published meta-analyses, PD-L1 expression in bladder cancer is related to advanced tumor stage and poor overall survival. However, conflicting results were demonstrated in some of the included studies and there were several confounding factors including patient characteristics, varying treatments, and definition of positivity. Thus, these associations should be interpreted with a caution [35, 36].

17.2.3. Dendritic Cells

Dendritic cells (DCs) are an important group of antigen presenting cells and are central regulators of the adaptive immune response in T cell-mediated anti-cancer immunity. Among DCs, a subset called conventional DCs (cDCs), especially cDC1s adept at inducing cellular immunity against cancer. They endocytose cancer cells or their debris and transport tumor-associated antigens (TAAs) to the draining lymph node, where cross-presentation of TAAs and priming of CD8+ T cells occurs [37, 38]. Mechanistic studies have identified several regulators of DC recruitment to the TME. CCL5 and XCL1 produced by natural killer cells have been found to recruit cDC1s to TME, which is impaired by tumor-derived pros-taglandin E2 [39]. Tumor-intrinsic signaling has also been linked to lack of priming of T cells by DCs. For example, in a melanoma mouse model, β-catenin signaling was found to inhibit CCL4 production and insufficient DCs recruitment and production of CXCL9 and CXCL10, which are required for effector T cell recruitment [40, 41]. The inverse correlation of β-catenin signaling and a T cell inflamed TME was subsequently demonstrated in human cancers including bladder cancer.

In human cancers expression of Batf3, a lineage marker of DCs necessary for T cell priming, was correlated with expression of CXCR3-binding chemokines (CXCL9, CXCL10, CXCL11) and CD8A expression [40]. In several types of cancers, it has been reported that patients with abundant infiltration of DCs in tumors by IHC or gene expression is correlated with better survival outcomes compared to patients with less DC infiltration [39, 4244]. In bladder cancer, DCs infiltration in tumors and DCs in urine have both been associated with efficacy of BCG therapy [45, 46]. We recently reported that CD8+ cells and Batf3+ DCs tended to spatially cluster rather than to distribute randomly, which suggests that those two types of immune cells cooperate in antitumor immunity (Fig. 17.2) [47].

Fig. 17.2.

Fig. 17.2

Multichannel immunofluorescence for CD8 and Batf3 markers on immune infiltrating cells from human bladder tissues showing clustering in a T cell-inflamed tumor (a and b), whereas a non-T cell-inflamed tumor (c and d) shows rare T cells without Batf3+ cells (60× magnification)

17.2.4. Regulatory T Cells

Regulatory T (Treg) cells are a subset of CD4+ T cells characterized by CD25 expression and the master transcription factor forkhead box protein P3 (FOXP3). These cells show immune-suppressive effects via various mechanism including cytotoxic T lymphocyte antigen 4 (CTLA-4)-mediated suppression of antigen-presenting cells, consumption of IL-2, and production of immune inhibitory cytokines and molecules [4850]. Although the molecular mechanism responsible for Treg expansion in TME is heterogeneous and remains uncertain, sphingosine 1 phosphate receptor 1 (S1P1) is overexpressed by bladder cancer cell and promotes the expression of TGF-β and IL-10 in vitro. [51] This process results in Treg expansion in bladder cancer.

Treg cells heavily infiltrate cancer tissue, and are absent or far less abundant in the peripheral circulation [50, 52]. This point highlights the importance of analyzing the TME. Similar to other cancers, human bladder cancer has abundant infiltration of Treg cells [5355]. Although abundant Treg cell infiltration has been reported to be associated with a poor prognosis, there have been several reports indicating the opposite association. For example, Treg infiltration is a favorable prognostic marker in colorectal cancer and Hodgkin’s lymphoma [5658]. In bladder cancer, while abundant infiltration of Treg cells was reported to be associated with a poor recurrence-free survival (RFS) or PFS after endoscopic resection [5961], the opposite relationship between Treg cell infiltration and survival outcome was also reported [62]. This paradoxical relationship between abundant Treg cell infiltration and favorable survival was explained by the suppression of matrix metalloproteinase 2 (MMP2) in TME, a key pro-invasive factor induced by tumor-promoting inflammation by Treg cells specifically at the invasive margin of the tumors [55]. In addition, several studies reported that high Treg cells/total T cells and high Treg cells/cytotoxic T cells were associated with poor survival outcomes. Also, the ratio of high cytotoxic T cells/Treg cells was associated with response to neoadjuvant chemotherapy [6365]. Therefore, the number of Treg cells is important, but in addition, the location and balance between Treg cells and other T cells are relevant. A more detailed classification of Treg cells including those with potential effector function might explain the discordant prognostic relevance in bladder cancer [66].

17.2.5. Tumor-Associated Macrophages

Tumor-associated macrophages (TAMs) are found in tumors that can suppress antitumor immunity and promote tumor growth and progression by producing cytokines and chemokines, promoting genetic instability, nurturing niche for cancer stem cells, and supporting metastasis [67]. TAMs are polarized to M1 or M2 according to signals from the TME and show different activities in terms of receptor expression, effector function, and cytokine and chemokine production. M1 macrophages are characterized by the production of pro-inflammatory and immune-stimulatory molecules, such as IL-12 and tumor necrosis factor alpha (TNF-α). M2 macrophages are characterized by the production of immune-suppressive molecules, such as IL-10, TGF-β, and IL-1ra [68]. TAMs are thought to be recruited from the blood compartment through chemotactic signals released from tumors, such as CCL2, CCL5, and CSF-1. In bladder cancer, CCL2 was also shown to recruit TAMs, which promoted lymphatic metastasis via vascular endothelial growth factor production [69].

In the TME, M2 immune-suppressive macrophages are predominant and M1 pro-inflammatory macrophages are relatively rare [67]. TAMs can be identified by expression of the surface marker, CD68. M2 macrophages can be identified using surface markers such as CD163 and CD204. Abundant infiltration of TAMs has been associated with high tumor grades and poor survival outcomes in various types of cancer [7075]. Interestingly, there are also some reports that describe discordant association between abundant infiltration of TAMs and survival outcomes according to treatments, suggesting that TAM infiltration is also related to efficacy of specific agents [76, 77]. In bladder cancer, abundant infiltration of TAMs, especially M2 phenotype, or a high M2/pan-macrophages ratio was linked to aggressiveness of the disease such as size, grade, nodal status, microvessels, and stage, as well as poor survival outcomes [7880]. These findings are consistent with the above-mentioned preclinical observations. Additionally, a high TAM/TIL ratio has been associated with a poor disease-free survival after cystectomy in patients with MIBC [81]. In patients treated with BCG after TURBT, abundant infiltration of TAMs or M2 macrophages was related to response to BCG and survival [45, 82, 83].

17.2.6. Myeloid-Derived Suppressor Cells

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid cells that strongly inhibit antitumor activities of other immune cells and also directly interact with tumor cells promoting tumor progression. MDSCs are not present in normal tissues, but appear in pathologic conditions such as cancer, chronic inflammation, infection, autoimmune diseases, trauma, and graft versus host disease. MDSCs are generally categorized into two phenotypes. Monocytic MDSCs (M-MDSCs) are phenotypically and morphologically similar to monocytes. Granulocytic or polymorphonuclear MDSCs (G-MDSCs or PMN-MDSCs) are phenotypically and morphologically similar to neutrophils. In humans, G-MDSCs and M-MDSCs are characterized by the markers CD11b+CD14CD15+ or CD11b+CD14CD66b+ and CD11b+CD14+HLA-DR−/loCD15, respectively [84, 85].

Evidence for the significance of MDSC has been accumulating in several cancers including bladder cancer. In vitro studies have revealed that G-MDSCs inhibit CD4+ T cell proliferation via CD15 and induce Treg cells [86]. Bladder cancer cells recruit MDSCs via CXCL2/MIF-CXCR2 signaling and also activate group 2 innate lymphoid cells (ILC2), which recruit and activate MDSCs further via IL-13 production [87, 88]. In relation to chemotherapy, cisplatin directly inhibited proliferation and induced the apoptosis using a human bladder cancer cell line, T24, in vitro [89]. Using a C3H/He mouse bladder cancer model, cisplatin treatment was also shown to inhibit the progression of the tumor and decrease the proportion of G-MDSCs phenotype in the tumor [89].

In human bladder cancers, the numbers of circulating G-MDSCs or total MDSCs are higher in bladder cancer patients than non-cancer healthy controls. In human bladder cancer tissue, G-MDSCs are the predominant phenotype and more G-MDSCs exist in cancer tissue than the adjacent normal tissue [86, 90, 91]. In addition, G-MDSCs are reported to be increased in cancer tissues compared to the adjacent non-cancer tissues, and a negative correlation was observed between the number of G-MDSCs and CD8+ T cells in tumors, which can be explained the expression of immune-suppressive molecules by MDSCs including Arg1, iNOS, ROS, PD-L1, and pSTAT3 [88, 89]. Increased MDSCs in peripheral blood or in bladder cancer tissue are associated with size, pathological grade, and stage of the tumor [88, 90, 91]. Patients with a low T cell/MDSC ratio in urine samples demonstrated a lower recurrence-free survival after BCG in patients with NMIBC. Patients with a high number of circulating MDSCs demonstrated a low pathological CR rate after neoadjuvant chemotherapy compared to patients with a low number of circulating MDSCs [87, 92].

17.2.7. Pericytes

Angiogenesis has been rigorously studied as a part of TME, since the aberrant vasculature is one of the main hallmarks of tumors, and suppresses oxygen distribution and immune cells delivery [93]. In angiogenesis, activation of tumor-associated endothelial cells was shown to promote tumor growth and progression in multiple human cancer cell lines, and a high inflammatory gene signature of the tumor-associated endothelial cells was related to a poor survival in gene expression analysis [94]. Meanwhile, pericytes, another component of small vessels, play important roles in angiogenesis by stabilizing capillaries and limiting vascular permeability [95]. Among two main subsets of pericytes in mice (type-1 [nestinNG2+] and type-2 [nestin+NG2+]), only type-2 pericytes were recruited during its angiogenesis in tumor plantation mouse models. In preclinical models, anti-angiogenetic agents showed increased pericyte coverage and enhances tumor perfusion and improved homogeneous oxygen distribution. Furthermore, it increased immune cells delivery and converted the TME from immunosuppressive state to immunostimulatory state [9698]. This phenomenon was also positively related to clinical outcomes in several clinical trials [98].

Following the success of combination of immune checkpoint inhibitors and axitinib as the first line therapy for advanced renal cell carcinoma [99, 100], the combination of an immune checkpoint inhibitor and an anti-angiogenetic agent is being tested for multiple cancer types including bladder cancer [97].

17.2.8. The Relationship Between the TME, Tumor-Cell Intrinsic Molecular Pathways, and the Responses to Immune Checkpoint Inhibitors

Although PD-L1 expression in TME by IHC was initially thought to be a promising predictive biomarker for response to anti-PD-1/L1 therapy, further research indicated that its accuracy remained limited [101]. Further research has focused on identifying other characteristics of the tumor microenvironment that might predict response. A biomarker analysis was performed on IMvigor210, a phase II trial of the anti-PD-L1 antibody atezolizumab in patients with metastatic urothelial cancer. The authors found that responses were associated with a CD8+ T-effector cell signature based on RNA sequencing and tumor mutation burden. Transforming growth factor β (TGF-β) signaling in fibroblasts predicted lack of response in patients with CD8+ T cell excluded tumors that had CD8+ T cell in peritumoral stroma [102]. More evidence about the relationship between the bladder cancer TME and responses to immune checkpoint inhibitor is expected, given the rapid accumulation data from bladder cancer patients receiving PD-1/PD-L1 treatment.

Based on genomic analyses of bladder cancer, several molecular subtypes have been proposed, and a recently proposed consensus molecular classification included six subtypes for muscle invasive bladder cancer [4, 7, 103]. Among them, “basal/squamous” tumors, which is the commonest subtype accounting for 35% of bladder cancer, were enriched in cytotoxic lymphocytes markers, natural killer cells markers, and monocytic lineage markers. This subtype was also associated with strong expression of immune checkpoint markers and antigen-presenting machinery genes, suggesting that such tumors might be more responsive to immunotherapies. The “stroma-rich” subtype overexpressed T cell markers and B cell markers [104]. Tumors with “luminal papillary” or “luminal unstable” classification lacked immune infiltrating cell markers as did the “neuroendocrine-like” subtype. “Luminal nonspecified” tumors showed limited expression for T and B cell markers and were associated with a fibroblastic stromal molecular pattern.

Evidence has accumulated that alterations in certain tumor-cell intrinsic molecular pathways are associated with the non-T cell-inflamed TME and resulting immunotherapy resistance. Gain-of-function alterations in Wnt-β catenin signaling and MYC and loss-of-function alterations or deletions LKB1, PTEN, and p53 were associated with reduced T cell priming or infiltration [105]. Activation of tumor-intrinsic Wnt-β catenin signaling was shown to be enriched in non-T cell-inflamed tumors across cancer types including bladder cancer using TCGA data [106]. Using the data of TCGA Bladder Urothelial Carcinoma, PPAR-γ, and FGFR3 pathways were activated in non-T cell-inflamed tumors as well as Wnt-β catenin signaling [29]. Indeed, activated PPARγ/RXRα signaling suppressed the production of pro-inflammatory cytokines and chemokines, resulting in impaired CD8+ T cell infiltration leading to resistance to immunotherapies in preclinical models [107]. FGFR3 mutation was associated with low T cell infiltration compared to wild type bladder cancers. The responsiveness to immunotherapy was not linked with FGFR alterations in the biomarker analyses from IMVIGOR 210 and Checkmate 275, which tested atezolizumab and nivolumab, respectively, in metastatic bladder cancer patients. It was suggested that an inverse association between FGFR3 mutation and a stromal TGF-β signaling was suggested to be the reason for similar response rates between FGFR3 mutated tumors and wild-type tumors, despite the difference of T cell infiltration [108].

17.3. Future Directions

The tumor microenvironment in bladder cancer is a complex of factors promoting and inhibiting the antitumor immune response. Therefore, a multidimensional approach to its evaluation will be necessary to gain a deeper understanding of the biological underpinnings at play. In addition to FACS or CyTOF, recently developed multiplex immunohistochemistry technology enabled us to stain multiple markers on a single slide and to evaluate multiple phenotypes of immune cells [109]. Besides quantitative analysis of the numbers of multiple phenotypes of infiltrating immune cells, spatial analysis can be conducted using this technology [47, 73]. Cytokines and chemokines also play important roles in terms of activation or inactivation and recruitment of immune cells, the relationships between these molecules and immune cells should be investigated for comprehensive understanding of TME. Combination of in situ hybridization for RNAs and immunohistochemistry for proteins could shed light on their relationships [110]. Emerging data indicate that heritable genetics and the commensal microbiome are two additional factors that can influence the tumor microenvironment in bladder cancer [111, 112]. There have been some reports suggesting interactions between nervous system and the tumor microenvironment and cancer progression [113117]. The roles of nerves impacting the TME in bladder cancer may be another important unexplored area of investigation. The incorporation of multiple interacting factors will necessitate the use of advanced statistical and computational approaches to characterize each unique tumor. These advances may enable us to better prevent, diagnose, prognosticate, and optimize treatments for bladder cancer patients in the future.

Acknowledgment

This work was supported by NIH K08CA234392, Cancer Research Foundation Young Investigator Award, and an Institutional Research Grant (#IRG-16-222-56) from the American Cancer Society and the Cancer Center Support Grant (#P30 CA14599) of the University of Chicago Medicine Comprehensive Cancer Center.

Declaration of Financial/Other Relationships:

R.F.S. reports consulting/honoraria from Aduro, AstraZeneca, BMS, Exelixis, Eisai, Mirati, Puma, and Medscape. R.F.S. reports research support (to institution) from Abbvie, Bayer, BMS, CytomX, Eisai, Genentech/Roche, Novartis, and Merck. K.H. reports fellowship funding from Japan Cancer Society. K.H. is currently a JSPS Overseas Research Fellow.

References

  • 1.Siegel RL, Miller KD, Jemal A (2018) Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30. Epub 2018/01/10. 10.3322/caac.21442 [DOI] [PubMed] [Google Scholar]
  • 2.Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386. Epub 2014/09/16. 10.1002/ijc.29210 [DOI] [PubMed] [Google Scholar]
  • 3.Moch H, Humphrey P, Ulbright T, Reuter V (2016) WHO classification of tumours of the urinary system and male genital organs-WHO classification of tumours, vol 8, 4th edn. IARC Press, Lyon [Google Scholar]
  • 4.(2014) Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 507(7492):315–322. 10.1038/nature12965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Choi W, Porten S, Kim S, Willis D, Plimack ER, Hoffman-Censits J, Roth B, Cheng T, Tran M, Lee IL, Melquist J, Bondaruk J, Majewski T, Zhang S, Pretzsch S, Baggerly K, Siefker-Radtke A, Czerniak B, Dinney CP, McConkey DJ (2014) Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 25(2):152–165. 10.1016/j.ccr.2014.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hedegaard J, Lamy P, Nordentoft I, Algaba F, Hoyer S, Ulhoi BP, Vang S, Reinert T, Hermann GG, Mogensen K, MBH T, Nielsen MM, Marquez M, Segersten U, Aine M, Hoglund M, Birkenkamp-Demtroder K, Fristrup N, Borre M, Hartmann A, Stohr R, Wach S, Keck B, Seitz AK, Nawroth R, Maurer T, Tulic C, Simic T, Junker K, Horstmann M, Harving N, Petersen AC, Calle ML, Steyerberg EW, Beukers W, van Kessel KEM, Jensen JB, Pedersen JS, Malmstrom PU, Malats N, Real FX, Zwarthoff EC, Orntoft TF, Dyrskjot L (2016) Comprehensive transcriptional analysis of early-stage urothelial carcinoma. Cancer Cell 30(1):27–42. 10.1016/j.ccell.2016.05.004. Epub 2016/06/21 [DOI] [PubMed] [Google Scholar]
  • 7.Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, Hinoue T, Laird PW, Hoadley KA, Akbani R, MAA C, Gibb EA, Kanchi RS, Gordenin DA, Shukla SA, Sanchez-Vega F, Hansel DE, Czerniak BA, Reuter VE, Su X, de Sa Carvalho B, Chagas VS, Mungall KL, Sadeghi S, Pedamallu CS, Lu Y, Klimczak LJ, Zhang J, Choo C, Ojesina AI, Bullman S, Leraas KM, Lichtenberg TM, Wu CJ, Schultz N, Getz G, Meyerson M, Mills GB, McConkey DJ, Weinstein JN, Kwiatkowski DJ, Lerner SP (2017) Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell 171(3):540–56.e25. 10.1016/j.cell.2017.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Martinez-Pineiro JA, Jimenez Leon J, Martinez-Pineiro L Jr, Fiter L, Mosteiro JA, Navarro J, Garcia Matres MJ, Carcamo P (1990) Bacillus Calmette-Guerin versus doxorubicin versus thiotepa: a randomized prospective study in 202 patients with superficial bladder cancer. J Urol 143(3):502–506. Epub 1990/03/01. 10.1016/s0022-5347(17)40002-4 [DOI] [PubMed] [Google Scholar]
  • 9.Bohle A, Bock PR (2004) Intravesical bacille Calmette-Guerin versus mitomycin C in superficial bladder cancer: formal meta-analysis of comparative studies on tumor progression. Urology 63(4):682–686.; Discussion 6–7. Epub 2004/04/10. 10.1016/j.urology.2003.11.049 [DOI] [PubMed] [Google Scholar]
  • 10.de Reijke TM, Kurth KH, Sylvester RJ, Hall RR, Brausi M, van de Beek K, Landsoght KE, Carpentier P (2005) Bacillus Calmette-Guerin versus epirubicin for primary, secondary or concurrent carcinoma in situ of the bladder: results of a European Organization for the Research and Treatment of Cancer-Genito-Urinary Group Phase III Trial (30906). J Urol 173(2):405–409. Epub 2005/01/12. 10.1097/01.ju.0000150425.09317.67 [DOI] [PubMed] [Google Scholar]
  • 11.Sylvester RJ, van der Meijden AP, Witjes JA, Kurth K (2005) Bacillus calmette-guerin versus chemotherapy for the intravesical treatment of patients with carcinoma in situ of the bladder: a meta-analysis of the published results of randomized clinical trials. J Urol 174(1):86–91. iscussion −2. Epub 2005/06/11. 10.1097/01.ju.0000162059.64886.1c [DOI] [PubMed] [Google Scholar]
  • 12.Mungan NA, Witjes JA (1998) Bacille Calmette-Guerin in superficial transitional cell carcinoma. Br J Urol 82(2):213–223. Epub 1998/09/02. 10.1046/j.1464-410x.1998.00720.x [DOI] [PubMed] [Google Scholar]
  • 13.Prescott S, Jackson AM, Hawkyard SJ, Alexandroff AB, James K (2000) Mechanisms of action of intravesical bacille Calmette-Guerin: local immune mechanisms. Clin Infect Dis 31(Suppl 3):S91–S93. Epub 2000/09/30. 10.1086/314066 [DOI] [PubMed] [Google Scholar]
  • 14.Bohle A, Brandau S (2003) Immune mechanisms in bacillus Calmette-Guerin immunotherapy for superficial bladder cancer. J Urol 170(3):964–969. Epub 2003/08/13. 10.1097/01.ju.0000073852.24341.4a [DOI] [PubMed] [Google Scholar]
  • 15.Redelman-Sidi G, Glickman MS, Bochner BH (2014) The mechanism of action of BCG therapy for bladder cancer-a current perspective. Nat Rev Urol 11(3):153–162. Epub 2014/02/05. 10.1038/nrurol.2014.15 [DOI] [PubMed] [Google Scholar]
  • 16.Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, Necchi A, Gerritsen W, Gurney H, Quinn DI, Culine S, Sternberg CN, Mai Y, Poehlein CH, Perini RF, Bajorin DF (2017) Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 376(11):1015–1026. 10.1056/NEJMoa1613683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Powles T, Duran I, van der Heijden MS, Loriot Y, Vogelzang NJ, De Giorgi U, Oudard S, Retz MM, Castellano D, Bamias A, Flechon A, Gravis G, Hussain S, Takano T, Leng N, Kadel EE 3rd, Banchereau R, Hegde PS, Mariathasan S, Cui N, Shen X, Derleth CL, Green MC, Ravaud A (2018) Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 391(10122):748–757. 10.1016/s0140-6736(17)33297-x. Epub 2017/12/23 [DOI] [PubMed] [Google Scholar]
  • 18.Apolo AB, Infante JR, Balmanoukian A, Patel MR, Wang D, Kelly K, Mega AE, Britten CD, Ravaud A, Mita AC, Safran H, Stinchcombe TE, Srdanov M, Gelb AB, Schlichting M, Chin K, Gulley JL (2017) Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, phase IB study. J Clin Oncol 35(19):2117–2124. 10.1200/jco.2016.71.6795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Powles T, O’Donnell PH, Massard C, Arkenau HT, Friedlander TW, Hoimes CJ, Lee JL, Ong M, Sridhar SS, Vogelzang NJ, Fishman MN, Zhang J, Srinivas S, Parikh J, Antal J, Jin X, Gupta AK, Ben Y, Hahn NM (2017) Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol 3(9):e172411. 10.1001/jamaoncol.2017.2411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, Plimack ER, Vaena D, Grimm MO, Bracarda S, Arranz JA, Pal S, Ohyama C, Saci A, Qu X, Lambert A, Krishnan S, Azrilevich A, Galsky MD (2017) Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 18(3):312–322. Epub 2017/01/31. 10.1016/s1470-2045(17)30065-7 [DOI] [PubMed] [Google Scholar]
  • 21.Balar AV, Castellano D, O’Donnell PH, Grivas P, Vuky J, Powles T, Plimack ER, Hahn NM, de Wit R, Pang L, Savage MJ, Perini RF, Keefe SM, Bajorin D, Bellmunt J (2017) First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 18(11):1483–1492. Epub 2017/10/03. 10.1016/s1470-2045(17)30616-2 [DOI] [PubMed] [Google Scholar]
  • 22.Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, Loriot Y, Necchi A, Hoffman-Censits J, Perez-Gracia JL, Dawson NA, van der Heijden MS, Dreicer R, Srinivas S, Retz MM, Joseph RW, Drakaki A, Vaishampayan UN, Sridhar SS, Quinn DI, Duran I, Shaffer DR, Eigl BJ, Grivas PD, Yu EY, Li S, Kadel EE 3rd, Boyd Z, Bourgon R, Hegde PS, Mariathasan S, Thastrom A, Abidoye OO, Fine GD, Bajorin DF (2017) Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 389(10064):67–76. 10.1016/s0140-6736(16)32455-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF (2009) Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res 69(7):3077–3085. 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12(4):252–264. 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zou W, Chen L (2008) Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 8(6):467–477. Epub 2008/05/27. 10.1038/nri2326 [DOI] [PubMed] [Google Scholar]
  • 26.Gajewski TF, Louahed J, Brichard VG (2010) Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy. Cancer J 16(4):399–403. 10.1097/PPO.0b013e3181eacbd8 [DOI] [PubMed] [Google Scholar]
  • 27.Senbabaoglu Y, Gejman RS, Winer AG, Liu M, Van Allen EM, de Velasco G, Miao D, Ostrovnaya I, Drill E, Luna A, Weinhold N, Lee W, Manley BJ, Khalil DN, Kaffenberger SD, Chen Y, Danilova L, Voss MH, Coleman JA, Russo P, Reuter VE, Chan TA, Cheng EH, Scheinberg DA, Li MO, Choueiri TK, Hsieh JJ, Sander C, Hakimi AA (2016) Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol 17(1):231. 10.1186/s13059-016-1092-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, Albright A, Cheng JD, Kang SP, Shankaran V, Piha-Paul SA, Yearley J, Seiwert TY, Ribas A, McClanahan TK (2017) IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127(8):2930–2940. 10.1172/jci91190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sweis RF, Spranger S, Bao R, Paner GP, Stadler WM, Steinberg G, Gajewski TF (2016) Molecular drivers of the non-t-cell-inflamed tumor microenvironment in urothelial bladder cancer. Cancer Immunol Res 4(7):563–568. 10.1158/2326-6066.CIR-15-0274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Faraj SF, Munari E, Guner G, Taube J, Anders R, Hicks J, Meeker A, Schoenberg M, Bivalacqua T, Drake C, Netto GJ (2015) Assessment of tumoral PD-L1 expression and intratumoral CD8+ T cells in urothelial carcinoma. Urology 85(3):703 e1–703 e6. 10.1016/j.urology.2014.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, Bajorin DF, Reuter VE, Herr H, Old LJ, Sato E (2007) CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma. Proc Natl Acad Sci U S A 104(10):3967–3972. 10.1073/pnas.0611618104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Krpina K, Babarovic E, Jonjic N (2015) Correlation of tumor-infiltrating lymphocytes with bladder cancer recurrence in patients with solitary low-grade urothelial carcinoma. Virchows Arch 467(4):443–448. Epub 2015/07/29. 10.1007/s00428-015-1808-6 [DOI] [PubMed] [Google Scholar]
  • 33.Wang B, Pan W, Yang M, Yang W, He W, Chen X, Bi J, Jiang N, Huang J, Lin T (2019) Programmed death ligand-1 is associated with tumor infiltrating lymphocytes and poorer survival in urothelial cell carcinoma of the bladder. Cancer Sci 110(2):489–498. 10.1111/cas.13887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hartana CA, Ahlen Bergman E, Broome A, Berglund S, Johansson M, Alamdari F, Jakubczyk T, Huge Y, Aljabery F, Palmqvist K, Holmstrom B, Glise H, Riklund K, Sherif A, Winqvist O (2018) Tissue-resident memory T cells are epigenetically cytotoxic with signs of exhaustion in human urinary bladder cancer. Clin Exp Immunol 194(1):39–53. 10.1111/cei.13183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ding X, Chen Q, Yang Z, Li J, Zhan H, Lu N, Chen M, Yang Y, Wang J, Yang D (2019) Clinicopathological and prognostic value of PD-L1 in urothelial carcinoma: a meta-analysis. Cancer Manag Res 11:4171–4184. 10.2147/cmar.S176937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zhu L, Sun J, Wang L, Li Z, Wang L, Li Z (2019) Prognostic and clinicopathological significance of PD-L1 in patients with bladder cancer: a meta-analysis. Front Pharmacol 10:962. 10.3389/fphar.2019.00962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gardner A, Ruffell B (2016) Dendritic cells and cancer immunity. Trends Immunol 37(12):855–865. 10.1016/j.it.2016.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF, Sancho D (2019) Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. Epub 2019/08/31. 10.1038/s41577-019-0210-z [DOI] [PubMed] [Google Scholar]
  • 39.Bottcher JP, Bonavita E, Chakravarty P, Blees H, Cabeza-Cabrerizo M, Sammicheli S, Rogers NC, Sahai E, Zelenay S, Reis e Sousa C (2018) NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 172(5):1022–1037.e14. 10.1016/j.cell.2018.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Spranger S, Dai D, Horton B, Gajewski TF (2017) Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31(5):711–23 e4. 10.1016/j.ccell.2017.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Spranger S, Bao R, Gajewski TF (2015) Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 523(7559):231–235. 10.1038/nature14404 [DOI] [PubMed] [Google Scholar]
  • 42.Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJ, Barczak A, Rosenblum MD, Daud A, Barber DL, Amigorena S, Van’t Veer LJ, Sperling AI, Wolf DM, Krummel MF (2014) Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26(5):638–652. 10.1016/j.ccell.2014.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Michea P, Noel F, Zakine E, Czerwinska U, Sirven P, Abouzid O, Goudot C, Scholer-Dahirel A, Vincent-Salomon A, Reyal F, Amigorena S, Guillot-Delost M, Segura E, Soumelis V (2018) Adjustment of dendritic cells to the breast-cancer microenvironment is subset specific. Nat Immunol 19(8):885–897. Epub 2018/07/18. 10.1038/s41590-018-0145-8 [DOI] [PubMed] [Google Scholar]
  • 44.Talmadge JE, Donkor M, Scholar E (2007) Inflammatory cell infiltration of tumors: Jekyll or Hyde. Cancer Metastasis Rev 26(3–4):373–400. Epub 2007/08/25. 10.1007/s10555-007-9072-0 [DOI] [PubMed] [Google Scholar]
  • 45.Ayari C, LaRue H, Hovington H, Decobert M, Harel F, Bergeron A, Tetu B, Lacombe L, Fradet Y (2009) Bladder tumor infiltrating mature dendritic cells and macrophages as predictors of response to bacillus Calmette-Guerin immunotherapy. Eur Urol 55(6):1386–1395. Epub 2009/02/06. 10.1016/j.eururo.2009.01.040 [DOI] [PubMed] [Google Scholar]
  • 46.Videira PA, Calais FM, Correia M, Ligeiro D, Crespo HJ, Calais F, Trindade H (2009) Efficacy of bacille Calmette-Guerin immunotherapy predicted by expression of antigen-presenting molecules and chemokines. Urology 74(4):944–950. Epub 2009/05/12. 10.1016/j.urology.2009.02.053 [DOI] [PubMed] [Google Scholar]
  • 47.Hatogai K, Kim D, Zha Y, Steinberg GD, Pearson AT, Gajewski TF, Sweis RF (2019) Multichannel immunofluorescence imaging to assess the immune composition of tumor microenvironment in bladder cancer. Cancer Res 79(13 Suppl) Abstract number 1093 [Google Scholar]
  • 48.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155(3):1151–1164. Epub 1995/08/01 [PubMed] [Google Scholar]
  • 49.Miyara M, Sakaguchi S (2011) Human FoxP3(+) CD4(+) regulatory T cells: their knowns and unknowns. Immunol Cell Biol 89(3):346–351. Epub 2011/02/09. 10.1038/icb.2010.137 [DOI] [PubMed] [Google Scholar]
  • 50.Togashi Y, Shitara K, Nishikawa H (2019) Regulatory T cells in cancer immunosuppression – implications for anticancer therapy. Nat Rev Clin Oncol 16(6):356–371. Epub 2019/02/02. 10.1038/s41571-019-0175-7 [DOI] [PubMed] [Google Scholar]
  • 51.Liu YN, Zhang H, Zhang L, Cai TT, Huang DJ, He J, Ni HH, Zhou FJ, Zhang XS, Li J (2019) Sphingosine 1 phosphate receptor-1 (S1P1) promotes tumor-associated regulatory T cell expansion: leading to poor survival in bladder cancer. Cell Death Dis 10(2):50. 10.1038/s41419-018-1298-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tada Y, Togashi Y, Kotani D, Kuwata T, Sato E, Kawazoe A, Doi T, Wada H, Nishikawa H, Shitara K (2018) Targeting VEGFR2 with Ramucirumab strongly impacts effector/ activated regulatory T cells and CD8(+) T cells in the tumor microenvironment. J Immunother Cancer 6(1):106. 10.1186/s40425-018-0403-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Loskog A, Ninalga C, Paul-Wetterberg G, de la Torre M, Malmstrom PU, Totterman TH (2007) Human bladder carcinoma is dominated by T-regulatory cells and Th1 inhibitory cytokines. J Urol 177(1):353–358. Epub 2006/12/13. 10.1016/j.juro.2006.08.078 [DOI] [PubMed] [Google Scholar]
  • 54.Zou W (2006) Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol 6(4):295–307. Epub 2006/03/25. 10.1038/nri1806 [DOI] [PubMed] [Google Scholar]
  • 55.Winerdal ME, Krantz D, Hartana CA, Zirakzadeh AA, Linton L, Bergman EA, Rosenblatt R, Vasko J, Alamdari F, Hansson J, Holmstrom B, Johansson M, Winerdal M, Marits P, Sherif A, Winqvist O (2018) Urinary bladder cancer tregs suppress MMP2 and potentially regulate invasiveness. Cancer Immunol Res 6(5):528–538. Epub 2018/03/29. 10.1158/2326-6066.Cir-17-0466 [DOI] [PubMed] [Google Scholar]
  • 56.Mirocha S, Elagin RB, Salamat S, Jaume JC (2009) T regulatory cells distinguish two types of primary hypophysitis. Clin Exp Immunol 155(3):403–411. 10.1111/j.1365-2249.2008.03828.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tanaka A, Sakaguchi S (2017) Regulatory T cells in cancer immunotherapy. Cell Res 27(1):109–118. 10.1038/cr.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Alvaro T, Lejeune M, Salvado MT, Bosch R, Garcia JF, Jaen J, Banham AH, Roncador G, Montalban C, Piris MA (2005) Outcome in Hodgkin’s lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res 11(4):1467–1473. Epub 2005/03/05. 10.1158/1078-0432.Ccr-04-1869 [DOI] [PubMed] [Google Scholar]
  • 59.Miyake M, Tatsumi Y, Gotoh D, Ohnishi S, Owari T, Iida K, Ohnishi K, Hori S, Morizawa Y, Itami Y, Nakai Y, Inoue T, Anai S, Torimoto K, Aoki K, Shimada K, Konishi N, Tanaka N, Fujimoto K (2017) Regulatory T cells and tumor-associated macrophages in the tumor microenvironment in non-muscle invasive bladder cancer treated with intravesical bacille calmette-guerin: a long-term follow-up study of a Japanese cohort. Int J Mol Sci 18(10). 10.3390/ijms18102186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Murai R, Itoh Y, Kageyama S, Nakayama M, Ishigaki H, Teramoto K, Narita M, Yoshida T, Tomita K, Kobayashi KI, Wada A, Nagasawa M, Kubota S, Ogasawara K, Kawauchi A (2018) Prediction of intravesical recurrence of non-muscle-invasive bladder cancer by evaluation of intratumoral Foxp3+ T cells in the primary transurethral resection of bladder tumor specimens. PLoS One 13(9):e0204745. 10.1371/journal.pone.0204745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tsai YS, Jou YC, Tung CL, Lin CT, Shen CH, Chen SY, Tsai HT, Lai CL, Wu CL, Tzai TS (2014) Loss of nuclear prothymosin-alpha expression is associated with disease progression in human superficial bladder cancer. Virchows Arch 464(6):717–724. Epub 2014/04/16. 10.1007/s00428-014-1578-6 [DOI] [PubMed] [Google Scholar]
  • 62.Winerdal ME, Marits P, Winerdal M, Hasan M, Rosenblatt R, Tolf A, Selling K, Sherif A, Winqvist O (2011) FOXP3 and survival in urinary bladder cancer. BJU Int 108(10):1672–1678. Epub 2011/01/20. 10.1111/j.1464-410X.2010.10020.x [DOI] [PubMed] [Google Scholar]
  • 63.Baras AS, Drake C, Liu JJ, Gandhi N, Kates M, Hoque MO, Meeker A, Hahn N, Taube JM, Schoenberg MP, Netto G, Bivalacqua TJ (2016) The ratio of CD8 to Treg tumor-infiltrating lymphocytes is associated with response to cisplatin-based neoadjuvant chemotherapy in patients with muscle invasive urothelial carcinoma of the bladder. Oncoimmunology 5(5):e1134412. 10.1080/2162402x.2015.1134412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Horn T, Laus J, Seitz AK, Maurer T, Schmid SC, Wolf P, Haller B, Winkler M, Retz M, Nawroth R, Gschwend JE, Kubler HR, Slotta-Huspenina J (2016) The prognostic effect of tumour-infiltrating lymphocytic subpopulations in bladder cancer. World J Urol 34(2):181–187. Epub 2015/06/10. 10.1007/s00345-015-1615-3 [DOI] [PubMed] [Google Scholar]
  • 65.Parodi A, Traverso P, Kalli F, Conteduca G, Tardito S, Curto M, Grillo F, Mastracci L, Bernardi C, Nasi G, Minaglia F, Simonato A, Carmignani G, Ferrera F, Fenoglio D, Filaci G (2016) Residual tumor micro-foci and overwhelming regulatory T lymphocyte infiltration are the causes of bladder cancer recurrence. Oncotarget 7(6):6424–6235. 10.18632/oncotarget.7024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, Mathian A, Nakahata T, Yamaguchi T, Nomura T, Ono M, Amoura Z, Gorochov G, Sakaguchi S (2009) Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30(6):899–911. [DOI] [PubMed] [Google Scholar]
  • 67.Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P (2017) Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14(7):399–416. 10.1038/nrclinonc.2016.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23(11):549–555. Epub 2002/10/29 [DOI] [PubMed] [Google Scholar]
  • 69.Chen C, He W, Huang J, Wang B, Li H, Cai Q, Su F, Bi J, Liu H, Zhang B, Jiang N, Zhong G, Zhao Y, Dong W, Lin T (2018) LNMAT1 promotes lymphatic metastasis of bladder cancer via CCL2 dependent macrophage recruitment. Nat Commun 9(1):3826. 10.1038/s41467-018-06152-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Atanasov G, Potner C, Aust G, Schierle K, Dietel C, Benzing C, Krenzien F, Bartels M, Eichfeld U, Schmelzle M, Bahra M, Pascher A, Wiltberger G (2018) TIE2-expressing monocytes and M2-polarized macrophages impact survival and correlate with angiogenesis in adenocarcinoma of the pancreas. Oncotarget 9(51):29715–29726. 10.18632/oncotarget.25690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hu Y, He MY, Zhu LF, Yang CC, Zhou ML, Wang Q, Zhang W, Zheng YY, Wang DM, Xu ZQ, Wu YN, Liu LK (2016) Tumor-associated macrophages correlate with the clinicopathological features and poor outcomes via inducing epithelial to mesenchymal transition in oral squamous cell carcinoma. J Exp Clin Cancer Res 35:12. 10.1186/s13046-015-0281-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zhang WJ, Wang XH, Gao ST, Chen C, Xu XY, Sun Q, Zhou ZH, Wu GZ, Yu Q, Xu G, Yao YZ, Guan WX (2018) Tumor-associated macrophages correlate with phenomenon of epithelial-mesenchymal transition and contribute to poor prognosis in triple-negative breast cancer patients. J Surg Res 222:93–101. Epub 2017/12/24. 10.1016/j.jss.2017.09.035 [DOI] [PubMed] [Google Scholar]
  • 73.Gartrell RD, Marks DK, Hart TD, Li G, Davari DR, Wu A, Blake Z, Lu Y, Askin KN, Monod A, Esancy CL, Stack EC, Jia DT, Armenta PM, Fu Y, Izaki D, Taback B, Rabadan R, Kaufman HL, Drake CG, Horst BA, Saenger YM (2018) Quantitative analysis of immune infiltrates in primary melanoma. Cancer Immunol Res 6(4):481–493. 10.1158/2326-6066.CIR-17-0360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sorensen MD, Dahlrot RH, Boldt HB, Hansen S, Kristensen BW (2018) Tumour-associated microglia/macrophages predict poor prognosis in high-grade gliomas and correlate with an aggressive tumour subtype. Neuropathol Appl Neurobiol 44(2):185–206. Epub 2017/08/03. 10.1111/nan.12428 [DOI] [PubMed] [Google Scholar]
  • 75.Tan KL, Scott DW, Hong F, Kahl BS, Fisher RI, Bartlett NL, Advani RH, Buckstein R, Rimsza LM, Connors JM, Steidl C, Gordon LI, Horning SJ, Gascoyne RD (2012) Tumor-associated macrophages predict inferior outcomes in classic Hodgkin lymphoma: a correlative study from the E2496 Intergroup trial. Blood 120(16):3280–3287. 10.1182/blood-2012-04-421057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Di Caro G, Cortese N, Castino GF, Grizzi F, Gavazzi F, Ridolfi C, Capretti G, Mineri R, Todoric J, Zerbi A, Allavena P, Mantovani A, Marchesi F (2016) Dual prognostic significance of tumour-associated macrophages in human pancreatic adenocarcinoma treated or untreated with chemotherapy. Gut 65(10):1710–1720. Epub 2015/07/15. 10.1136/gutjnl-2015-309193 [DOI] [PubMed] [Google Scholar]
  • 77.Taskinen M, Karjalainen-Lindsberg ML, Nyman H, Eerola LM, Leppa S (2007) A high tumor-associated macrophage content predicts favorable outcome in follicular lymphoma patients treated with rituximab and cyclophosphamide-doxorubicin-vincristine-prednisone. Clin Cancer Res 13(19):5784–5789. Epub 2007/10/03. 10.1158/1078-0432.Ccr-07-0778 [DOI] [PubMed] [Google Scholar]
  • 78.Hanada T, Nakagawa M, Emoto A, Nomura T, Nasu N, Nomura Y (2000) Prognostic value of tumor-associated macrophage count in human bladder cancer. Int J Urol 7(7):263–269. Epub 2000/07/26. 10.1046/j.1442-2042.2000.00190.x [DOI] [PubMed] [Google Scholar]
  • 79.Takeuchi H, Tanaka M, Tanaka A, Tsunemi A, Yamamoto H (2016) Predominance of M2-polarized macrophages in bladder cancer affects angiogenesis, tumor grade and invasiveness. Oncol Lett 11(5):3403–3408. 10.3892/ol.2016.4392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wang B, Liu H, Dong X, Wu S, Zeng H, Liu Z, Wan D, Dong W, He W, Chen X, Zheng L, Huang J, Lin T (2015) High CD204+ tumor-infiltrating macrophage density predicts a poor prognosis in patients with urothelial cell carcinoma of the bladder. Oncotarget 6(24):20204–20214. 10.18632/onco-target.3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sjodahl G, Lovgren K, Lauss M, Chebil G, Patschan O, Gudjonsson S, Mansson W, Ferno M, Leandersson K, Lindgren D, Liedberg F, Hoglund M (2014) Infiltration of CD3(+) and CD68(+) cells in bladder cancer is subtype specific and affects the outcome of patients with muscle-invasive tumors. Urol Oncol 32(6):791–797. Epub 2014/05/06. 10.1016/j.urolonc.2014.02.007 [DOI] [PubMed] [Google Scholar]
  • 82.Lima L, Oliveira D, Tavares A, Amaro T, Cruz R, Oliveira MJ, Ferreira JA, Santos L (2014) The predominance of M2-polarized macrophages in the stroma of low-hypoxic bladder tumors is associated with BCG immunotherapy failure. Urol Oncol 32(4):449–457. Epub 2013/11/21. 10.1016/j.urolonc.2013.10.012 [DOI] [PubMed] [Google Scholar]
  • 83.Suriano F, Santini D, Perrone G, Amato M, Vincenzi B, Tonini G, Muda A, Boggia S, Buscarini M, Pantano F (2013) Tumor associated macrophages polarization dictates the efficacy of BCG instillation in non-muscle invasive urothelial bladder cancer. J Exp Clin Cancer Res:32–87. 10.1186/1756-9966-32-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gabrilovich DI (2017) Myeloid-derived suppressor cells. Cancer Immunol Res 5(1):3–8. 10.1158/2326-6066.Cir-16-0297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yin Z, Li C, Wang J, Xue L (2019) Myeloid-derived suppressor cells: roles in the tumor microenvironment and tumor radiotherapy. Int J Cancer 144(5):933–946. Epub 2018/07/12. 10.1002/ijc.31744 [DOI] [PubMed] [Google Scholar]
  • 86.Eruslanov E, Neuberger M, Daurkin I, Perrin GQ, Algood C, Dahm P, Rosser C, Vieweg J, Gilbert SM, Kusmartsev S (2012) Circulating and tumor-infiltrating myeloid cell subsets in patients with bladder cancer. Int J Cancer 130(5):1109–1119. Epub 2011/04/12. 10.1002/ijc.26123 [DOI] [PubMed] [Google Scholar]
  • 87.Chevalier MF, Trabanelli S, Racle J, Salome B, Cesson V, Gharbi D, Bohner P, Domingos-Pereira S, Dartiguenave F, Fritschi AS, Speiser DE, Rentsch CA, Gfeller D, Jichlinski P, Nardelli-Haefliger D, Jandus C, Derre L (2017) ILC2-modulated T cell-to-MDSC balance is associated with bladder cancer recurrence. J Clin Invest 127(8):2916–2929. 10.1172/jci89717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang H, Ye YL, Li MX, Ye SB, Huang WR, Cai TT, He J, Peng JY, Duan TH, Cui J, Zhang XS, Zhou FJ, Wang RF, Li J (2017) CXCL2/MIF-CXCR2 signaling promotes the recruitment of myeloid-derived suppressor cells and is correlated with prognosis in bladder cancer. Oncogene 36(15):2095–2104. Epub 2016/10/11. 10.1038/onc.2016.367 [DOI] [PubMed] [Google Scholar]
  • 89.Wu K, Tan MY, Jiang JT, Mu XY, Wang JR, Zhou WJ, Wang X, Li MQ, He YY, Liu ZH (2018) Cisplatin inhibits the progression of bladder cancer by selectively depleting G-MDSCs: a novel chemoimmunomodulating strategy. Clin Immunol 193:60–69. Epub 2018/02/08. 10.1016/j.clim.2018.01.012 [DOI] [PubMed] [Google Scholar]
  • 90.Yang G, Shen W, Zhang Y, Liu M, Zhang L, Liu Q, Lu HH, Bo J (2017) Accumulation of myeloid-derived suppressor cells (MDSCs) induced by low levels of IL-6 correlates with poor prognosis in bladder cancer. Oncotarget 8(24):38378–38388. 10.18632/oncotarget.16386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yuan XK, Zhao XK, Xia YC, Zhu X, Xiao P (2011) Increased circulating immunosuppressive CD14(+)HLA-DR(−/low) cells correlate with clinical cancer stage and pathological grade in patients with bladder carcinoma. J Int Med Res 39(4):1381–1391. Epub 2011/10/12. 10.1177/147323001103900424 [DOI] [PubMed] [Google Scholar]
  • 92.Ornstein MC, Diaz-Montero CM, Rayman P, Elson P, Haywood S, Finke JH, Kim JS, Pavicic PG Jr, Lamenza M, Devonshire S, Dann P, Schach K, Stephenson A, Campbell S, Emamekhoo H, Ernstoff MS, Hoimes CJ, Gilligan TD, Rini BI, Garcia JA, Grivas P (2018) Myeloid-derived suppressors cells (MDSC) correlate with clinicopathologic factors and pathologic complete response (pCR) in patients with urothelial carcinoma (UC) undergoing cystectomy. Urol Oncol 36(9):405–412. 10.1016/j.urolonc.2018.02.018. Epub 2018/04/03 [DOI] [PubMed] [Google Scholar]
  • 93.De Palma M, Biziato D, Petrova TV (2017) Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer 17(8):457–474. Epub 2017/07/15. 10.1038/nrc.2017.51 [DOI] [PubMed] [Google Scholar]
  • 94.Pitroda SP, Zhou T, Sweis RF, Filippo M, Labay E, Beckett MA, Mauceri HJ, Liang H, Darga TE, Perakis S, Khan SA, Sutton HG, Zhang W, Khodarev NN, Garcia JG, Weichselbaum RR (2012) Tumor endothelial inflammation predicts clinical outcome in diverse human cancers. PLoS One 7(10):e46104. 10.1371/journal.pone.0046104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Armulik A, Genove G, Betsholtz C (2011) Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 21(2):193–215. Epub 2011/08/16. 10.1016/j.devcel.2011.07.001 [DOI] [PubMed] [Google Scholar]
  • 96.Huang Y, Yuan J, Righi E, Kamoun WS, Ancukiewicz M, Nezivar J, Santosuosso M, Martin JD, Martin MR, Vianello F, Leblanc P, Munn LL, Huang P, Duda DG, Fukumura D, Jain RK, Poznansky MC (2012) Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A 109(43):17561–17566. 10.1073/pnas.1215397109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ciciola P, Cascetta P, Bianco C, Formisano L, Bianco R (2020) Combining immune checkpoint inhibitors with anti-angiogenic agents. J Clin Med 9(3). 10.3390/jcm9030675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Jain RK (2014) Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 26(5):605–622. 10.1016/j.ccell.2014.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, Lee JL, Vasiliev A, Miller WH Jr, Gurney H, Schmidinger M, Larkin J, Atkins MB, Bedke J, Alekseev B, Wang J, Mariani M, Robbins PB, Chudnovsky A, Fowst C, Hariharan S, Huang B, di Pietro A, Choueiri TK (2019) Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med 380(12):1103–1115. 10.1056/NEJMoa1816047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulieres D, Melichar B, Vynnychenko I, Kryzhanivska A, Bondarenko I, Azevedo SJ, Borchiellini D, Szczylik C, Markus M, McDermott RS, Bedke J, Tartas S, Chang YH, Tamada S, Shou Q, Perini RF, Chen M, Atkins MB, Powles T (2019) Pembrolizumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med 380(12):1116–1127. Epub 2019/02/20. 10.1056/NEJMoa1816714 [DOI] [PubMed] [Google Scholar]
  • 101.Ghate K, Amir E, Kuksis M, Hernandez-Barajas D, Rodriguez-Romo L, Booth CM, Vera-Badillo FE (2019) PD-L1 expression and clinical outcomes in patients with advanced urothelial carcinoma treated with checkpoint inhibitors: a meta-analysis. Cancer Treat Rev 76:51–56. Epub 2019/05/28. 10.1016/j.ctrv.2019.05.002 [DOI] [PubMed] [Google Scholar]
  • 102.Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, Kadel EE III, Koeppen H, Astarita JL, Cubas R, Jhunjhunwala S, Banchereau R, Yang Y, Guan Y, Chalouni C, Ziai J, Senbabaoglu Y, Santoro S, Sheinson D, Hung J, Giltnane JM, Pierce AA, Mesh K, Lianoglou S, Riegler J, Carano RAD, Eriksson P, Hoglund M, Somarriba L, Halligan DL, van der Heijden MS, Loriot Y, Rosenberg JE, Fong L, Mellman I, Chen DS, Green M, Derleth C, Fine GD, Hegde PS, Bourgon R, Powles T (2018) TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554(7693):544–548. 10.1038/nature25501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kamoun A, de Reynies A, Allory Y, Sjodahl G, Robertson AG, Seiler R, Hoadley KA, Groeneveld CS, Al-Ahmadie H, Choi W, Castro MAA, Fontugne J, Eriksson P, Mo Q, Kardos J, Zlotta A, Hartmann A, Dinney CP, Bellmunt J, Powles T, Malats N, Chan KS, Kim WY, McConkey DJ, Black PC, Dyrskjot L, Hoglund M, Lerner SP, Real FX, Radvanyi F (2019) A consensus molecular classification of muscle-invasive bladder cancer. Eur Urol. Epub 2019/09/30. 10.1016/j.eururo.2019.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kamoun A, Reynies A, Allory Y, Sjodahl G, Robertson AG, Seiler R, Hoadley KA, Groeneveld CS, Al-Ahmadie H, Choi W, Castro MAA, Fontugne J, Eriksson P, Mo Q, Kardos J, Zlotta A, Hartmann A, Dinney CP, Bellmunt J, Powles T, Malats N, Chan KS, Kim WY, McConkey DJ, Black PC, Dyrskjot L, Hoglund M, Lerner SP, Real FX, Radvanyi F (2019) Bladder cancer molecular taxonomy G. A consensus molecular classification of muscle-invasive bladder cancer. Eur Urol. Epub 2019/09/30. 10.1016/j.eururo.2019.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Spranger S, Gajewski TF (2018) Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer 18(3):139–147. Epub 2018/01/13. 10.1038/nrc.2017.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Luke JJ, Bao R, Sweis RF, Spranger S, Gajewski TF (2019) WNT/beta-catenin pathway activation correlates with immune exclusion across human cancers. Clin Cancer Res 25(10):3074–3083. 10.1158/1078-0432.CCR-18-1942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Korpal M, Puyang X, Jeremy Wu Z, Seiler R, Furman C, Oo HZ, Seiler M, Irwin S, Subramanian V, Julie Joshi J, Wang CK, Rimkunas V, Tortora D, Yang H, Kumar N, Kuznetsov G, Matijevic M, Chow J, Kumar P, Zou J, Feala J, Corson L, Henry R, Selvaraj A, Davis A, Bloudoff K, Douglas J, Kiss B, Roberts M, Fazli L, Black PC, Fekkes P, Smith PG, Warmuth M, Yu L, Hao MH, Larsen N, Daugaard M, Zhu P (2017) Evasion of immunosurveillance by genomic alterations of PPARgamma/RXRalpha in bladder cancer. Nat Commun 8(1):103. 10.1038/s41467-017-00147-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wang L, Gong Y, Saci A, Szabo PM, Martini A, Necchi A, Siefker-Radtke A, Pal S, Plimack ER, Sfakianos JP, Bhardwaj N, Horowitz A, Farkas AM, Mulholland D, Fischer BS, Oh WK, Sharma P, Zhu J, Galsky MD (2019) Fibroblast growth factor receptor 3 alterations and response to PD-1/PD-L1 blockade in patients with metastatic urothelial cancer. Eur Urol 76(5):599–603. 10.1016/j.eururo.2019.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Stack EC, Wang C, Roman KA, Hoyt CC (2014) Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods 70(1):46–58. Epub 2014/09/23. 10.1016/j.ymeth.2014.08.016 [DOI] [PubMed] [Google Scholar]
  • 110.Wee YTF, Alkaff SMF, Lim JCT, Loh JJH, Hilmy MH, Ong C, Nei WL, Jain A, Lim A, Takano A, Azhar R, Wan WK, Newell E, Yeong J, Lim TKH (2018) An integrated automated multispectral imaging technique that simultaneously detects and quantitates viral RNA and immune cell protein markers in fixed sections from Epstein-Barr virus-related tumours. Ann Diagn Pathol 37:12–19. Epub 2018/09/16. 10.1016/j.anndiagpath.2018.09.002 [DOI] [PubMed] [Google Scholar]
  • 111.Sayaman RW, Saad M, Thorsson V, Hendrickx W, Roelands J, Mokrab Y, Farshidfar F, Kirchhoff T, Sweis RF, Bathe OF, Porta-Pardo E, Campbell MJ, Stretch C, Hu D, Huntsman S, Graff RE, Syed N, Radvanyi L, Shelley S, Wolf D, Marincola FM, Ceccarelli M, Galon J, Ziv E, Bedognetti D (2020) Germline genetic contribution to the immune landscape of cancer. 10.1101/2020.01.30.926527. bioRxiv. 2020.01.30.926527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sweis RF, Golan S, Barashi N, Hill E, Andolfi C, Werntz RP, Bloodworth J, Steinberg GD (2019) Association of the commensal urinary microbiome with response to Bacillus Calmette-Guérin (BCG) immunotherapy in non-muscle invasive bladder cancer. J Clin Oncol 37(suppl 7S; abstr 423) [Google Scholar]
  • 113.Magnon C, Hall SJ, Lin J, Xue X, Gerber L, Freedland SJ, Frenette PS (2013) Autonomic nerve development contributes to prostate cancer progression. Science 341(6142):1236361. Epub 2013/07/13. 10.1126/science.1236361 [DOI] [PubMed] [Google Scholar]
  • 114.Pedro HDM, Prazeres CL, Silva WN, Santos GSP, Costa AC, Picoli CC, Gonçalves WA, Vieira MS, Costa PAC, Rocha BGS, Sena IFG, Campos LMCC, Paz MT, Costa MR, Resende RR, Cunha TM, Mintz A, Birbrair A (2020) Ablation of sensory nerves favours melanoma progression. J Cell Mol Med [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zahalka AH, Arnal-Estapé A, Maryanovich M, Nakahara F, Cruz CD, Finley LWS, Frenette PS (2017) Adrenergic nerves activate an angio-metabolic switch in prostate cancer. Science 358(6361):321–326. 10.1126/science.aah5072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hayakawa Y, Sakitani K, Konishi M, Asfaha S, Niikura R, Tomita H, Renz BW, Tailor Y, Macchini M, Middelhoff M, Jiang Z, Tanaka T, Dubeykovskaya ZA, Kim W, Chen X, Urbanska AM, Nagar K, Westphalen CB, Quante M, Lin CS, Gershon MD, Hara A, Zhao CM, Chen D, Worthley DL, Koike K, Wang TC (2017) Nerve growth factor promotes gastric tumorigenesis through aberrant cholinergic signaling. Cancer Cell 31(1):21–34. 10.1016/j.ccell.2016.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ayala GE, Wheeler TM, Shine HD, Schmelz M, Frolov A, Chakraborty S, Rowley D (2001) In vitro dorsal root ganglia and human prostate cell line interaction: redefining perineural invasion in prostate cancer. Prostate 49(3):213–223. Epub 2001/12/18. 10.1002/pros.1137 [DOI] [PubMed] [Google Scholar]

RESOURCES