Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.401
Filter
1.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38847806

ABSTRACT

Due to bladder tumors' contact with urine, urine-derived cells (UDCs) may serve as a surrogate for monitoring the tumor microenvironment (TME) in bladder cancer (BC). However, the composition of UDCs and the extent to which they mirror the tumor remain poorly characterized. We generated the first single-cell RNA-sequencing of BC patient UDCs with matched tumor and peripheral blood mononuclear cells (PBMC). BC urine was more cellular than healthy donor (HD) urine, containing multiple immune populations including myeloid cells, CD4+ and CD8+ T cells, natural killer (NK) cells, B cells, and dendritic cells (DCs) in addition to tumor and stromal cells. Immune UDCs were transcriptionally more similar to tumor than blood. UDCs encompassed cytotoxic and activated CD4+ T cells, exhausted and tissue-resident memory CD8+ T cells, macrophages, germinal-center-like B cells, tissue-resident and adaptive NK cells, and regulatory DCs found in tumor but lacking or absent in blood. Our findings suggest BC UDCs may be surrogates for the TME and serve as therapeutic biomarkers.


Subject(s)
Tumor Microenvironment , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Humans , Tumor Microenvironment/immunology , Male , Killer Cells, Natural/immunology , Female , CD8-Positive T-Lymphocytes/immunology , Aged , CD4-Positive T-Lymphocytes/immunology , Single-Cell Analysis/methods , Dendritic Cells/immunology , Middle Aged , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , RNA-Seq , Single-Cell Gene Expression Analysis
2.
Investig Clin Urol ; 65(3): 263-278, 2024 May.
Article in English | MEDLINE | ID: mdl-38714517

ABSTRACT

PURPOSE: Myofibroblastic cancer-associated fibroblasts (myCAFs) are important components of the tumor microenvironment closely associated with tumor stromal remodeling and immunosuppression. This study aimed to explore myCAFs marker gene biomarkers for clinical diagnosis and therapy for patients with bladder cancer (BC). MATERIALS AND METHODS: BC single-cell RNA sequencing (scRNA-seq) data were obtained from the National Center for Biotechnology Information Sequence Read Archive. Transcriptome and clinical data were downloaded from The Cancer Genome Atlas and the Gene Expression Omnibus databases. Subsequently, univariate Cox and LASSO (Least Absolute Shrinkage and Selection Operator regression) regression analyses were performed to construct a prognostic signature. Immune cell activity was estimated using single-sample gene set enrichment analysis whilst the TIDE (tumor immune dysfunction and exclusion) method was employed to assess patient response to immunotherapy. The chemotherapy response of patients with BC was evaluated using genomics of drug sensitivity in cancer. Furthermore, Immunohistochemistry was used to verify the correlation between MAP1B expression and immunotherapy efficacy. The scRNA-seq data were analyzed to identify myCAFs marker genes. RESULTS: Combined with bulk RNA-sequencing data, we constructed a two-gene (COL6A1 and MAP1B) risk signature. In patients with BC, the signature demonstrated outstanding prognostic value, immune infiltration, and immunotherapy response. This signature served as a crucial guide for the selection of anti-tumor chemotherapy medications. Additionally, immunohistochemistry confirmed that MAP1B expression was significantly correlated with immunotherapy efficacy. CONCLUSIONS: Our findings revealed a typical prognostic signature based on myCAF marker genes, which offers patients with BC a novel treatment target alongside theoretical justification.


Subject(s)
Biomarkers, Tumor , Cancer-Associated Fibroblasts , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/drug therapy , Prognosis , Biomarkers, Tumor/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Male , Female , Transcriptome , Treatment Outcome , Myofibroblasts
3.
Proc Natl Acad Sci U S A ; 121(20): e2312855121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713626

ABSTRACT

The immune landscape of bladder cancer progression is not fully understood, and effective therapies are lacking in advanced bladder cancer. Here, we visualized that bladder cancer cells recruited neutrophils by secreting interleukin-8 (IL-8); in turn, neutrophils played dual functions in bladder cancer, including hepatocyte growth factor (HGF) release and CCL3highPD-L1high super-immunosuppressive subset formation. Mechanistically, c-Fos was identified as the mediator of HGF up-regulating IL-8 transcription in bladder cancer cells, which was central to the positive feedback of neutrophil recruitment. Clinically, compared with serum IL-8, urine IL-8 was a better biomarker for bladder cancer prognosis and clinical benefit of immune checkpoint blockade (ICB). Additionally, targeting neutrophils or hepatocyte growth factor receptor (MET) signaling combined with ICB inhibited bladder cancer progression and boosted the antitumor effect of CD8+ T cells in mice. These findings reveal the mechanism by which tumor-neutrophil cross talk orchestrates the bladder cancer microenvironment and provide combination strategies, which may have broad impacts on patients suffering from malignancies enriched with neutrophils.


Subject(s)
Disease Progression , Interleukin-8 , Neutrophils , Tumor Microenvironment , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/immunology , Tumor Microenvironment/immunology , Humans , Neutrophils/immunology , Neutrophils/metabolism , Animals , Mice , Interleukin-8/metabolism , Cell Line, Tumor , Hepatocyte Growth Factor/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Female , Male , Neutrophil Infiltration
4.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38731992

ABSTRACT

Non-muscle-invasive papillary urothelial carcinoma (NMIPUC) of the urinary bladder is the most common type of bladder cancer. Intravesical Bacille Calmette-Guerin (BCG) immunotherapy is applied in patients with a high risk of recurrence and progression of NMIPUC to muscle-invasive disease. However, the tumor relapses in about 30% of patients despite the treatment, raising the need for better risk stratification. We explored the potential of spatial distributions of immune cell subtypes (CD20, CD11c, CD163, ICOS, and CD8) within the tumor microenvironment to predict NMIPUC recurrence following BCG immunotherapy. Based on analyses of digital whole-slide images, we assessed the densities of the immune cells in the epithelial-stromal interface zone compartments and their distribution, represented by an epithelial-stromal interface density ratio (IDR). While the densities of any cell type did not predict recurrence, a higher IDR of CD11c (HR: 0.0012, p-value = 0.0002), CD8 (HR: 0.0379, p-value = 0.005), and ICOS (HR: 0.0768, p-value = 0.0388) was associated with longer recurrence-free survival (RFS) based on the univariate Cox regression. The history of positive repeated TUR (re-TUR) (HR: 4.93, p-value = 0.0001) and T1 tumor stage (HR: 2.04, p-value = 0.0159) were associated with shorter RFS, while G3 tumor grade according to the 1973 WHO classification showed borderline significance (HR: 1.83, p-value = 0.0522). In a multivariate analysis, the two models with a concordance index exceeding 0.7 included the CD11c IDR in combination with either a history of positive re-TUR or tumor stage. We conclude that the CD11c IDR is the most informative predictor of NMIPUC recurrence after BCG immunotherapy. Our findings highlight the importance of assessment of the spatial distribution of immune cells in the tumor microenvironment.


Subject(s)
BCG Vaccine , Immunotherapy , Macrophages , Neoplasm Recurrence, Local , Tumor Microenvironment , Urinary Bladder Neoplasms , Humans , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Male , BCG Vaccine/therapeutic use , Neoplasm Recurrence, Local/immunology , Female , Immunotherapy/methods , Aged , Middle Aged , Macrophages/immunology , Macrophages/metabolism , Carcinoma, Papillary/pathology , Carcinoma, Papillary/immunology , Carcinoma, Papillary/therapy , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Prognosis , Aged, 80 and over
5.
Nat Commun ; 15(1): 4448, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789460
6.
Aging (Albany NY) ; 16(9): 7774-7798, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38696324

ABSTRACT

BACKGROUND: Dysregulation of the immune system and N6-methyladenosine (m6A) contribute to immune therapy resistance and cancer progression in urothelial carcinoma (UC). This study aims to identify immune-related molecules, that are m6A-modified, and that are associated with tumor progression, poor prognosis, and immunotherapy response. METHODS: We identified prognostic immune genes (PIGs) using Cox analysis and random survival forest variable hunting algorithm (RSF-VH) on immune genes retrieved from the Immunology Database and Analysis Portal database (ImmPort). The RM2Target database and MeRIP-seq analysis, combined with a hypergeometric test, assessed m6A methylation in these PIGs. We analyzed the correlation between the immune pattern and prognosis, as well as their association with clinical factors in multiple datasets. Moreover, we explored the interplay between immune patterns, tumor immune cell infiltration, and m6A regulators. RESULTS: 28 PIGs were identified, of which the 10 most significant were termed methylated prognostic immune genes (MPIGs). These MPIGs were used to create an immune pattern score. Kaplan-Meier and Cox analyses indicated this pattern as an independent risk factor for UC. We observed significant associations between the immune pattern, tumor progression, and immune cell infiltration. Differential expression analysis showed correlations with m6A regulators expression. This immune pattern proved effective in predicting immunotherapy response in UC in real-world settings. CONCLUSION: The study identified a m6A-modified immune pattern in UC, offering prognostic and therapeutic response predictions. This emphasizes that immune genes may influence tumor immune status and progression through m6A modifications.


Subject(s)
Adenosine , Immunotherapy , Humans , Adenosine/analogs & derivatives , Prognosis , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/therapy
7.
Front Immunol ; 15: 1360436, 2024.
Article in English | MEDLINE | ID: mdl-38812516

ABSTRACT

Bladder cancer is a common type of cancer around the world, and the majority of patients are diagnosed with non-muscle-invasive bladder cancer (NMIBC). Although low-risk NMIBC has a good prognosis, the disease recurrence rate and development of treatment-refractory disease remain high in intermediate- to high-risk NMIBC patients. To address these challenges for the treatment of NMIBC, a novel combination therapy composed of an oncolytic adenovirus (oAd) co-expressing interleukin (IL)-12, granulocyte-macrophage colony-stimulating factor (GM-CSF), and relaxin (RLX; HY-oAd) and a clinical-stage glycogen synthase kinase (GSK)-3ß inhibitor (9-ING-41; elraglusib) was investigated in the present report. Our findings demonstrate that HY-oAd and 9-ING-41 combination therapy (HY-oAd+9-ING-41) exerted superior inhibition of tumor growth compared with respective monotherapy in a syngeneic NMIBC tumor model. HY-oAd+9-ING-41 induced high-level tumor extracellular matrix (ECM) degradation and a more potent antitumor immune response than the respective monotherapy. In detail, HY-oAd+9-ING-41 induced superior accumulation of intratumoral T cells, prevention of immune cell exhaustion, and induction of tumor-specific adaptive immune response compared to either monotherapy. Collectively, these results demonstrate that the combination of HY-oAd and 9-ING-41 may be a promising approach to elicit a potent antitumor immune response against bladder cancer.


Subject(s)
Adenoviridae , Glycogen Synthase Kinase 3 beta , Oncolytic Virotherapy , Oncolytic Viruses , Tumor Microenvironment , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Animals , Adenoviridae/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Mice , Humans , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Cell Line, Tumor , Combined Modality Therapy , Female
8.
J Cell Mol Med ; 28(10): e18384, 2024 May.
Article in English | MEDLINE | ID: mdl-38760964

ABSTRACT

Smoking is a well-known risk factor for non-small-cell lung cancer (NSCLC) and bladder urothelial carcinoma (BLCA). Despite this, there has been no investigation into a prognostic marker based on smoking-related genes that could universally predict prognosis in these cancers and correlate with immune checkpoint therapy. This study aimed to identify smoking-related differential genes in NSCLC and BLCA, analyse their roles in patient prognosis and immune checkpoint therapy through subgroup analyses, and shed light on PRR11 as a crucial prognostic gene in both cancers. By examining PRR11 co-expressed genes, a prognostic model was constructed and its impact on immunotherapy for NSCLC and BLCA was evaluated. Molecular docking and tissue microarray analyses were conducted to explore the correlation between PRR11 and its reciprocal gene SPDL1. Additionally, miRNAs associated with PRR11 were analysed. The study confirmed a strong link between smoking-related genes, prognosis, and immune checkpoint therapy in NSCLC and BLCA. PRR11 was identified as a key smoking-associated gene that influences the efficacy of immune checkpoint therapy by modulating the stemness of these cancers. A prognostic model based on PRR11 co-expressed genes in BLCA was established and its prognostic value was validated in NSCLC. Furthermore, it was found that PRR11 regulates PDL1 via SPDL1, impacting immunotherapeutic efficacy in both cancers. The involvement of hsa-miR-200b-3p in the regulation of SPDL1 expression by PRR11 was also highlighted. Overall, the study elucidates that PRR11 modulates patient immunotherapy by influencing PDL1 expression through its interaction with SPDL1, with potential upstream regulation by hsa-miR-200b-3p.


Subject(s)
Gene Expression Regulation, Neoplastic , Immunotherapy , Lung Neoplasms , MicroRNAs , Smoking , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Prognosis , Smoking/adverse effects , Immunotherapy/methods , MicroRNAs/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Male , Female
9.
Int Immunopharmacol ; 133: 111892, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38663315

ABSTRACT

BACKGROUND: Bladder cancer (BC), a prevalent malignancy in the urinary system, often poses challenges for effective treatment. Immunotherapy, harnessing the immune system, has exhibited promise in early-stage clinical trials. Mucosal associated invariant T (MAIT) cells, a subset of immune cells implicated in various diseases, including certain cancer, have yet to be explored in BC patients. We aimed to investigate the quantity, function, and anti-tumor effects of MAIT cells in BC patients. METHODS: A total of 75 newly diagnosed BC patients and 183 healthy volunteers were included. Blood samples were collected and analyzed to evaluate the quantity and function of MAIT cells. Surgical resection provided BC tissues for further analysis, and the clinical features of BC tumors were collected and their relationship with MAIT cells was explored. RESULTS: MAIT cells were identified in both healthy individuals and BC patients. The proportion of MAIT cells in the peripheral blood of BC patients did not significantly differ from that of healthy controls. However, the study revealed a correlation between the proportion of IFN-γ producing MAIT cells and tumor number and invasion in BC patients. Furthermore, MAIT cells exhibited cytotoxic effects on BC cells in vitro and in vivo. CONCLUSIONS: This study sheds light on the role of MAIT cells in BC. While the quantity of MAIT cells showed no significant change in BC patients, their functional attributes and association with tumor characteristics suggest their potential as an immunotherapy target in BC treatment.


Subject(s)
Mucosal-Associated Invariant T Cells , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Mucosal-Associated Invariant T Cells/immunology , Male , Female , Middle Aged , Aged , Animals , Interferon-gamma/metabolism , Adult , Cell Line, Tumor , Immunotherapy/methods , Mice , Cytotoxicity, Immunologic
10.
Cells ; 13(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38667314

ABSTRACT

The clinical management of bladder cancer continues to present significant challenges. Bacillus Calmette-Guérin (BCG) immunotherapy remains the gold standard of treatment for non-muscle invasive bladder cancer (NMIBC), but many patients develop recurrence and progression to muscle-invasive disease (MIBC), which is resistant to BCG. This review focuses on the immune mechanisms mobilized by BCG in bladder cancer tumor microenvironments (TME), mechanisms of BCG resistance, the dual role of the BCG-triggered NFkB/TNFα/PGE2 axis in the regulation of anti-tumor and tumor-promoting aspects of inflammation, and emerging strategies to modulate their balance. A better understanding of BCG resistance will help develop new treatments and predictive biomarkers, paving the way for improved clinical outcomes in bladder cancer patients.


Subject(s)
BCG Vaccine , Immunotherapy , Tumor Microenvironment , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Humans , Tumor Microenvironment/immunology , BCG Vaccine/therapeutic use , BCG Vaccine/immunology , Immunotherapy/methods , Animals
11.
Curr Opin Urol ; 34(4): 244-250, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38630912

ABSTRACT

PURPOSE OF REVIEW: Bladder cancer (BC) is a highly heterogenous disease comprising tumours of various molecular subtypes and histologic variants. This heterogeneity represents a major challenge for the development of novel therapeutics. Preclinical models that closely mimic in vivo tumours and reflect their diverse biology are indispensable for the identification of therapies with specific activity in various BC subtypes. In this review, we summarize efforts and progress made in this context during the last 24 months. RECENT FINDINGS: In recent years, one main focus was laid on the development of patient-derived BC models. Patient-derived organoids (PDOs) and patient-derived xenografts (PDXs) were demonstrated to widely recapitulate the molecular and histopathological characteristics, as well as the drug response profiles of the corresponding tumours of origin. These models, thus, represent promising tools for drug development and personalized medicine. Besides PDXs, syngenic in vivo models are of growing importance. Since these models are generated using immunocompetent hosts, they can, amongst others, be used to develop novel immunotherapeutics and to evaluate the impact of the immune system on drug response and resistance. SUMMARY: In the past two years, various in vivo and in vitro models closely recapitulating the biology and heterogeneity of human bladder tumours were developed.


Subject(s)
Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Humans , Animals , Disease Models, Animal , Organoids , Precision Medicine/methods , Xenograft Model Antitumor Assays , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
12.
Curr Opin Urol ; 34(4): 236-243, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38650456

ABSTRACT

PURPOSE OF REVIEW: Current risk stratification and treatment decision-making for bladder cancer informed by histopathology as well as molecular diagnostics face limitations. This review summarizes recent advancements in single-cell and spatial omics methodologies for understanding bladder cancer biology and their potential impact on development of novel therapeutic strategies. RECENT FINDINGS: Single-cell RNA sequencing and spatial omics techniques offer unprecedented insights into various aspects of tumor microenvironment (TME), bladder cancer heterogeneity, cancer stemness, and cellular plasticity. Studies have identified multiple malignant cell subpopulations within tumors, revealing diverse transcriptional states and clonal evolution. Additionally, intratumor heterogeneity has been linked to tumor progression and therapeutic response. Immune cell composition analysis has revealed immunosuppressive features in the TME, impacting treatment response. Furthermore, studies have elucidated the role of cancer-associated fibroblasts and endothelial cells in shaping the tumor immune landscape and response to therapy. SUMMARY: Single-cell and spatial omics technologies have revolutionized our understanding of bladder cancer biology, uncovering previously unseen complexities. These methodologies provide valuable insights into tumor heterogeneity and microenvironmental interactions, with implications for therapeutic development. However, challenges remain in translating research findings into clinical practice and implementing personalized treatment strategies. Continued interdisciplinary collaboration and innovation are essential for overcoming these challenges and leveraging the full potential of single-cell and spatial omics in improving bladder cancer diagnosis and treatment.


Subject(s)
Single-Cell Analysis , Tumor Microenvironment , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Single-Cell Analysis/methods , Tumor Microenvironment/immunology
13.
Toxicology ; 504: 153782, 2024 May.
Article in English | MEDLINE | ID: mdl-38493947

ABSTRACT

Tobacco carcinogens are recognized as critical hazard factors for bladder tumorigenesis, affecting the prognosis of patients through aromatic amines components. However, the specific function of tobacco carcinogens and systematic assessment models in the prognosis of bladder cancer remains poorly elucidated. We retrieved bladder cancer specific tobacco carcinogens-related genes from Comparative Toxicogenomic Database, our Nanjing Bladder Cancer cohort and TCGA database. Gene×Gene interaction method was utilized to establish a prognostic signature. Integrative assessment of immunogenomics, tumor microenvironments and single-cell RNA-sequencing were performed to illustrate the internal relations of key events from different levels. Finally, we comprehensively identified 33 essential tobacco carcinogens-related genes to construct a novel prognostic signature, and found that high-risk patients were characterized by significantly worse overall survival (HR=2.25; Plog-rank < 0.01). Single-cell RNA-sequencing and multi-omics analysis demonstrated that cancer-associated fibroblasts mediated the crosstalk between epithelial-mesenchymal transition progression and immune evasion. Moreover, an adverse outcome pathway framework was established to facilitate our understanding to the tobacco carcinogens-triggered bladder tumorigenesis. Our study systematically provided immune microenvironmental alternations for smoking-induced adverse survival outcomes in bladder cancer. These findings facilitated the integrative multi-omics insights into risk assessment and toxic mechanisms of tobacco carcinogens.


Subject(s)
Cancer-Associated Fibroblasts , Epithelial-Mesenchymal Transition , Tumor Microenvironment , Urinary Bladder Neoplasms , Humans , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/drug effects , Carcinogens/toxicity , Gene Expression Regulation, Neoplastic , Immune Evasion , Multiomics , Prognosis , Single-Cell Analysis , Smoking/adverse effects , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
14.
Cancer Biomark ; 40(1): 79-94, 2024.
Article in English | MEDLINE | ID: mdl-38517777

ABSTRACT

BACKGROUND: N6-methyladenosine (m6A) is the most frequent RNA modification in mammals, and its role in bladder cancer (BC) remains rarely revealed. OBJECTIVE: To predict the value of m6A-related genes in prognosis and immunity in BC. METHODS: We performed multiple omics analysis of 618 TCGA and GEO patients and used principal component analysis (PCA) to calculate the m6A score for BC patients. RESULTS: We described the multiple omics status of 23 m6A methylation-related genes (MRGs), and four m6A clusters were identified, which showed significant differences in immune infiltration and biological pathways. Next, we intersected the differential genes among m6A clusters, and 11 survival-related genes were identified, which were used to calculate the m6A score for the patients. We found that the high-score (HS) group showed lower tumor mutation burden (TMB) and TP53 mutations and better prognosis than the low-score (LS) group. Lower immune infiltration, higher expression of PD-L1, PD-1, and CTLA4, and higher immune dysfunction and immune exclusion scores were identified in the LS group, suggesting a higher possibility of immune escape. Finally, the experimental verification shows that the m6A related genes, such as IGFBP1, plays an important role in the growth and metastasis of bladder cancer. CONCLUSIONS: These findings revealed the important roles of m6A MRGs in predicting prognosis, TMB status, TP53 mutation, immune functions and immunotherapeutic response in BC.


Subject(s)
Adenosine , Biomarkers, Tumor , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/mortality , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics , Prognosis , Biomarkers, Tumor/genetics , Tumor Escape/genetics , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Multiomics
15.
Int J Cancer ; 155(2): 352-364, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38483404

ABSTRACT

Treatment for higher-risk non-muscle invasive bladder cancer (NMIBC) involves intravesical immunotherapy with Bacillus Calmette Guérin (BCG); however, disease recurrence and progression occur frequently. Systemic immunity is critical for successful cancer immunotherapy; thus, recurrence of NMIBC may be due to suboptimal systemic activation of anti-tumor immunity after local immunotherapy. We previously reported that systemically acquired trained immunity (a form of innate immune memory) in circulating monocytes is associated with increased time-to-recurrence in patients with NMIBC treated with BCG. Herein, we used a mouse model of NMIBC to compare the effects of intravesical versus intravenous (systemic) BCG immunotherapy on the local and peripheral immune microenvironments. We also assessed whether BCG-induced trained immunity modulates anti-tumor immune responses. Compared with intravesical BCG, which led to a tumor-promoting immune microenvironment, intravenous BCG resulted in an anti-tumoral bladder microenvironment characterized by increased proportions of cytotoxic T lymphocytes (CTLs), and decreased proportions of myeloid-derived suppressor cells. Polarization toward anti-tumoral immunity occurred in draining lymph nodes, spleen, and bone marrow following intravenous versus intravesical BCG treatment. Pre-treatment with intravesical BCG was associated with increased rate of tumor growth compared with intravenous BCG pre-treatment. Trained immunity contributed to remodeling of the tumor immune microenvironment, as co-instillation of BCG-trained macrophages with ovalbumin-expressing bladder tumor cells increased the proportion of tumor-specific CTLs. Furthermore, BCG-trained dendritic cells exhibited enhanced antigen uptake and presentation and promoted CTL proliferation. Our data support the concept that systemic immune activation promotes anti-tumor responses, and that BCG-induced trained immunity is important in driving anti-tumor adaptive immunity.


Subject(s)
BCG Vaccine , Immunotherapy , Tumor Microenvironment , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/drug therapy , Animals , Tumor Microenvironment/immunology , Mice , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , BCG Vaccine/therapeutic use , Immunotherapy/methods , Female , Administration, Intravesical , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Humans , Disease Models, Animal , Immunity, Innate/immunology , Cell Line, Tumor , Immunologic Memory/immunology , Myeloid-Derived Suppressor Cells/immunology , Trained Immunity
16.
Altern Ther Health Med ; 30(4): 130-138, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38518167

ABSTRACT

Objective: The emergence of immunotherapy has heralded a profound transformation in the therapeutic landscape of bladder cancer (BLAC). Immunotherapy, with its unique potential for "combination therapy", has brought about greater possibilities for treating BLCA. However, there is significant heterogeneity among bladder cancer patients, and a portion of those in advanced stages may not experience substantial benefits from chemotherapy. Immunotherapy offers a potential ray of hope for specific patient subsets. Thus, predicting the effectiveness of tumor immunotherapy and providing them with more precise treatment strategies hold paramount importance and clinical value in delivering personalized therapeutic interventions for advanced bladder cancer patients. This study is designed to establish a risk score model derived from immune-related genes that can effectively assess prognosis and immunotherapy outcomes in patients with bladder cancer. Methods: The IMvigor210 dataset served as our training set for developing the prognostic model based on immune-related genes. Robust 7-gene expression patterns were investigated from the training set. A time-dependent receiver operating characteristic (ROC) curve and Kaplan-Meier (KM)analysis were employed to determine the prognostic relevance of these gene patterns. Independent datasets collected from the Cancer Genome Atlas Program (TCGA) and Gene Expression Omnibus (GEO) databases were additionally utilized for re-determination. The association between the 7-gene signature-based risk score and immunological subtypes, tumor mutational burden (TMB), immune checkpoint expressions, and the proportion of immune cell infiltration was assessed within training and test sets. Furthermore, the training set's predictive potential for immunotherapy response was assessed using the 7-gene signature, and its validity was externally verified on three datasets (GSE176307, GSE140901, and GSE91016). By validating the 7-gene signature externally, we eneralized the findings beyond the original training set, and assessed the model's performance in diverse contexts. Consistent performance across these datasets reinforces the robustness and clinical utility of our 7-gene signature. Results: Employing the transcriptional and clinical information from the IMvigor210 for training, 348 patients were classified into two clusters with notable distinctions in prognostic stratification and immunotherapy efficacy. Seven immune-related genes Indoleamine 2,3-dioxygenase 1 (IDO1), TNF receptor superfamily member 17 (TNFRSF17), Killer Cell Lectin Like Receptor K1 (KLRK1), TNF receptor superfamily member 14 (TNFSF14), Lymphocyte-activation gene 3 (LAG3), Killer Cell Lectin Like Receptor C1 (KLRC1), and Ecto-5'-nucleotidase (NT5E) were screened based on different expression genes (DEGs) between the two clusters. The expression levels of these seven genes and the accompanying univariate component Cox regression coefficients, were computed to create a 7-gene signature-based risk score. The median value of the risk score was utilized to categorize the BLCA individuals into high-risk and low-risk groups. Researchers identified that in the low-risk group, individuals exhibited a noticeably improved chance of surviving. The external validation cohorts verified the risk score model's prognostic capacity. Furthermore, it was demonstrated that while low-risk individuals possessed higher TMB scores, higher expression of immune checkpoint genes, and lower levels of immunological infiltration, they responded more favorably to immunotherapy. The clinical relevance of the risk score model was validated in three immunotherapy groups. Conclusion: The risk score model might be utilized to forecast the prognosis and efficacy of immunotherapy in BLCA patients, offering a novel course of treatment for these individuals. For patients undergoing immunotherapy, this gene signature can help predict treatment response. Low-risk patients may benefit from more tailored monitoring and personalized immunotherapy regimens. However, more investigations are required to validate its accuracy and effectiveness in a prospective cohort with larger sample sizes.


Subject(s)
Immunotherapy , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Humans , Prognosis , Immunotherapy/methods , Biomarkers, Tumor/genetics , Female , Male
17.
Int J Biol Markers ; 39(2): 158-167, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38321777

ABSTRACT

BACKGROUND: Typically, the overexpressed keratin 7 (KRT7) is considered a validated therapeutic target and prognosis marker in bladder cancer. However, the crucial roles of KRT7 in the clinical prognosis and immune microenvironment in bladder cancer remain unclear. METHODS: Initially, the expression levels of KRT7 in public databases were analyzed that is,Tumor Immune Estimation Resource (TIMER) 2.0 and Gene Expression Profiling Interactive Analysis (GEPIA). Further, the clinical tissue samples from patients (n = 10 pairs) were collected to confirm the expression trends of KRT7 and detected by immunohistochemistry (IHC) analysis. Meanwhile, the relationship between KRT7 and the prognosis of bladder cancer patients was analyzed by Kaplan-Meier plotter estimation and Cox regression analysis. Finally, TIMER 2.0 and IHC staining analyses were performed to calculate the infiltration abundances of three kinds of immune cells in eligible bladder tumor samples. RESULTS: The TIMER 2.0 and GEPIA datasets suggested the differences in the expression levels of KRT7 in tumors, in which KRT7 was significantly upregulated in bladder cancer. The KRT7 expression was closely associated with patients' gender, tumor histologic subtypes, T status, and American Joint Committee on Cancer stages. Notably, the increased KRT7 indicated poor overall survival and disease-free survival rates. Moreover, KRT7 expression could be responsible for immune infiltration in the cancer microenvironment of the bladder. Finally, the high expression level of KRT7 increased the presence of regulatory T cells (Tregs) but reduced the infiltration of CD8+ T and natural killer cells. CONCLUSION: KRT7 as a biomarker potentiated the prediction of bladder cancer prognosis and the immune microenvironment.


Subject(s)
Keratin-7 , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/immunology , Prognosis , Keratin-7/metabolism , Keratin-7/genetics , Female , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Male , Tumor Microenvironment , Middle Aged , Clinical Relevance
19.
Cancer Sci ; 115(5): 1417-1432, 2024 May.
Article in English | MEDLINE | ID: mdl-38422408

ABSTRACT

Platelets and M2 macrophages both play crucial roles in tumorigenesis, but their relationship and the prognosis value of the relative genes in bladder cancer (BLCA) remain obscure. In the present study, we found that platelets stimulated by BLCA cell lines could promote M2 macrophage polarization, and platelets were significantly associated with the infiltration of M2 macrophages in BLCA samples. Through the bioinformatic analyses, A2M, TGFB3, and MYLK, which were associated with platelets and M2 macrophages, were identified and verified in vitro and then included in the predictive model. A platelet and M2 macrophage-related gene signature was constructed to evaluate the prognosis and immunotherapeutic sensitivity, helping to guide personalized treatment and to disclose the underlying mechanisms.


Subject(s)
Blood Platelets , Immunotherapy , Macrophages , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Humans , Prognosis , Macrophages/immunology , Macrophages/metabolism , Blood Platelets/metabolism , Cell Line, Tumor , Immunotherapy/methods , Male , Female , Gene Expression Regulation, Neoplastic , Computational Biology/methods , Mice , Transcriptome , Middle Aged , Gene Expression Profiling/methods
20.
J Pathol ; 263(2): 139-149, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38380548

ABSTRACT

TP53 mutation is one of the most common genetic alterations in urothelial carcinoma (UrCa), and heterogeneity of TP53 mutants leads to heterogeneous clinical outcomes. This study aimed to investigate the clinical relevance of specific TP53 mutations in UrCa. In this study, a total of eight cohorts were enrolled, along with matched clinical annotation. TP53 mutations were classified as disruptive and nondisruptive according to the degree of disturbance of p53 protein function and structure. We evaluated the clinical significance of TP53 mutations in our local datasets and publicly available datasets. The co-occurring events of TP53 mutations in UrCa, along with their therapeutic indications, functional effects, and the tumor immune microenvironment, were also investigated. TP53 mutations were identified in 49.7% of the UrCa patients. Within this group, 25.1% of patients carried TP53Disruptive mutations, a genetic alteration correlated with a significantly poorer overall survival (OS) when compared to individuals with TP53Nondisruptive mutations and those with wild-type TP53. Significantly, patients with TP53Disruptive mutations exhibit an increased probability of responding favorably to PD-1/PD-L1 blockade and chemoimmunotherapy. Meanwhile, there was no noteworthy distinction in OS among patients with varying TP53 mutation status who underwent chemotherapy. Samples with TP53Disruptive mutations showed an enriched APOBEC- and POLE-related mutational signature, as well as an elevated tumor mutation burden. The sensitivity to immunotherapy in tumors carrying TP53Disruptive mutation may be attributed to the inflamed tumor microenvironment characterized by increased CD8+T cell infiltration and interferon-gamma signaling activation. In conclusion, UrCa patients with TP53Disruptive mutations have shown reduced survival rates, yet they may respond well to PD-1/PD-L1 blockade therapy and chemoimmunotherapy. By distinguishing specific TP53 mutations, we can improve risk stratification and offer personalized genomics-guided therapy to UrCa patients. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Mutation , Programmed Cell Death 1 Receptor , Tumor Microenvironment , Tumor Suppressor Protein p53 , Urinary Bladder Neoplasms , Humans , Tumor Suppressor Protein p53/genetics , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/immunology , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/mortality , Biomarkers, Tumor/genetics , Male , Female , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aged , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...