1
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
2
|
Regulatory T Cells but Not Tumour-Infiltrating Lymphocytes Correlate with Tumour Invasion Depth in Basal Cell Carcinoma. Diagnostics (Basel) 2022; 12:diagnostics12122987. [PMID: 36552993 PMCID: PMC9776706 DOI: 10.3390/diagnostics12122987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Basal cell carcinoma (BCC) is the most common skin malignancy worldwide. Current evidence suggests tumour-infiltrating lymphocytes (TILs) may influence the clinical outcomes of patients with BCC. The present study aimed to profile the infiltrative characteristics of stromal TILs and regulatory T cells (Treg cells) in the tumour centre (TC), tumour periphery (TP), and normal adjacent tissue (NAT) of BCC. A total of 111 samples from 43 cutaneous BCC cases were examined for TIL (CD3+) and Treg cell (FOXP3+/CD3+) expression using immunohistochemical techniques. The correlations of Treg cells with TILs, invasion depth, and tumour morphological risk were analysed. We identified a high mean proportion of Treg cells within the tumour (TC = 46.9%, TP = 56.1%, NAT = 51.8%) despite a relatively low median of TILs (TC = 12.7%, TP = 10.3%, NAT = 3.6%), supporting the classification of BCC as a cold tumour. A significant positive correlation was observed between the proportion of Treg cells and sTILs (ρ = 0.325, p < 0.001), suggesting a predominant role of TILs in the infiltration of Treg cells. An inverse correlation discovered between Treg cells and tumour invasion depth (r = −0.36, p = 0.017) might indicate Treg cells’ anti-tumour capacity in BCC.
Collapse
|
3
|
Bieri U, Enderlin D, Buser L, Wettstein MS, Eberli D, Moch H, Hermanns T, Poyet C. Modified immunoscore improves the prediction of progression-free survival in patients with non-muscle-invasive bladder cancer: A digital pathology study. Front Oncol 2022; 12:964672. [PMID: 36212478 PMCID: PMC9539272 DOI: 10.3389/fonc.2022.964672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour-infiltrating lymphocytes (TIL), known to be of prognostic value in various solid tumours, have been in the focus of research in the last years. TIL are often quantified via IMMUNOSCORE ® (IS), a scoring system based on TIL cell densities. Recent studies were able to replicate these findings for muscle-invasive bladder cancer (MIBC), however data regarding non-muscle-invasive bladder cancer (NMIBC) are scarce. This study aimed to evaluate the value of a modified Immunoscore (mIS) as a predictive marker for NMIBC prognosis using tissue-micro-arrays (TMAs). We analysed two TMAs containing 316 samples from 158 patients with NMIBC, stained for CD3, CD8, CD45RO and FOXP3. Stained TIL were captured by digital pathology, cumulated, averaged, and reported as density (stained cells per mm²). The mIS was then constructed based on density of all four immune-cell types. Clinical, pathological and follow-up data were collected retrospectively. Univariable and multivariable cox regression analysis was performed to assess the potential value of mIS as a predictor for progression free survival (PFS) and recurrence-free-survival (RFS). Patients within "European Organisation for Research and Treatment of Cancer" (EORTC) risk groups were further substratified in high mIS and low mIS subgroups. Finally log-rank test was used to compare the different survival curves. The median age in our cohort was 68 years (Interquartile Range (IQR): 60 - 76), and 117 (74%) patients were male. A total of 26 patients (16.5%) were classified as EORTC low risk, 45 (28.5%) as intermediate risk and 87 (55.1%) as high risk. Patients in the EORTC high risk group with low mIS showed a shorter PFS in comparison to high mIS (HR 2.9, CI 0.79 - 11.0, p=0.082). In contrast, no predictive potential regarding PFS was observed in intermediate or low risk groups. Furthermore, mIS was not able to predict RFS in any EORTC risk group. mIS could be utilized to predict prognosis more accurately in high-risk patients with NMIBC by identifying those with higher or lower risk of progression. Therefore, mIS could be used to allocate these highrisk patients to more streamlined follow-up or more aggressive treatment strategies.
Collapse
Affiliation(s)
- Uwe Bieri
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Dominik Enderlin
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Lorenz Buser
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Marian S. Wettstein
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Cédric Poyet
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Lombardo KA, Obradovic A, Singh AK, Liu JL, Joice G, Kates M, Bishai W, McConkey D, Chaux A, Eich ML, Rezaei MK, Netto GJ, Drake CG, Tran P, Matoso A, Bivalacqua TJ. BCG invokes superior STING-mediated innate immune response over radiotherapy in a carcinogen murine model of urothelial cancer. J Pathol 2022; 256:223-234. [PMID: 34731491 PMCID: PMC8738146 DOI: 10.1002/path.5830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/07/2021] [Accepted: 10/30/2021] [Indexed: 02/03/2023]
Abstract
Radiation and bacillus Calmette-Guérin (BCG) instillations are used clinically for treatment of urothelial carcinoma, but the precise mechanisms by which they activate an immune response remain elusive. The role of the cGAS-STING pathway has been implicated in both BCG and radiation-induced immune response; however, comparison of STING pathway molecules and the immune landscape following treatment in urothelial carcinoma has not been performed. We therefore comprehensively analyzed the local immune response in the bladder tumor microenvironment following radiotherapy and BCG instillations in a well-established spontaneous murine model of urothelial carcinoma to provide insight into activation of STING-mediated immune response. Mice were exposed to the oral carcinogen, BBN, for 12 weeks prior to treatment with a single 15 Gy dose of radiation or three intravesical instillations of BCG (1 × 108 CFU). At sacrifice, tumors were staged by a urologic pathologist and effects of therapy on the immune microenvironment were measured using the NanoString Myeloid Innate Immunity Panel and immunohistochemistry. Clinical relevance was established by measuring immune biomarker expression of cGAS and STING on a human tissue microarray consisting of BCG-treated non-muscle-invasive urothelial carcinomas. BCG instillations in the murine model elevated STING and downstream STING-induced interferon and pro-inflammatory molecules, intratumoral M1 macrophage and T-cell accumulation, and complete tumor eradication. In contrast, radiotherapy caused no changes in STING pathway or innate immune gene expression; rather, it induced M2 macrophage accumulation and elevated FoxP3 expression characteristic of immunosuppression. In human non-muscle-invasive bladder cancer, STING protein expression was elevated at baseline in patients who responded to BCG therapy and increased further after BCG therapy. Overall, these results show that STING pathway activation plays a key role in effective BCG-induced immune response and strongly indicate that the effects of BCG on the bladder cancer immune microenvironment are more beneficial than those induced by radiation. © 2021 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kara A Lombardo
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Greenberg Bladder Cancer Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Aleksandar Obradovic
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Alok Kumar Singh
- Center for Tuberculosis Research, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - James L Liu
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Gregory Joice
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Max Kates
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - William Bishai
- Center for Tuberculosis Research, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - David McConkey
- Greenberg Bladder Cancer Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Alcides Chaux
- Department of Scientific Research, School of Postgraduate Studies, Norte University, 1614 Asunción, Paraguay
| | - Marie-Lisa Eich
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - M Katayoon Rezaei
- Department of Pathology, George Washington University, Washington, DC, USA
| | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles G Drake
- Division of Urology, Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Division Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Phuoc Tran
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Radiation Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Andres Matoso
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Greenberg Bladder Cancer Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Trinity J Bivalacqua
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Greenberg Bladder Cancer Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
5
|
Pinard CJ, Stegelmeier AA, Bridle BW, Mutsaers AJ, Wood RD, Wood GA, Woods JP, Hocker SE. Evaluation of lymphocyte-specific programmed cell death protein 1 receptor expression and cytokines in blood and urine in canine urothelial carcinoma patients. Vet Comp Oncol 2021; 20:427-436. [PMID: 34797014 DOI: 10.1111/vco.12788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/04/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022]
Abstract
Urothelial carcinoma (UC) is the most common urinary tumour in dogs. Despite a range of treatment options, prognosis remains poor in dogs. In people, breakthroughs with checkpoint inhibitors have established new standards of care for muscle-invasive bladder cancer patients and elevated levels of programmed cell death protein 1 (PD-1) suggest immune checkpoint blockade may be a novel target for therapy. The goal of this study was to determine if canine UC patients express elevated levels of lymphocyte-specific PD-1 and/or urinary cytokine biomarkers compared to healthy dogs. Paired blood and urine were evaluated in 10 canine UC patients, five cystitis patients and 10 control dogs for lymphocyte-specific PD-1 expression via flow cytometry and relative cytokine expression. In UC patients, PD-1 expression was significantly elevated on CD8+ lymphocytes in urine samples. UC patients had a higher CD4:CD8 ratio in their urine compared to healthy dogs, however, there was no significant variation in the CD8:Treg ratio between any group. Cystitis patients had significantly elevated levels of CD4+ T cells, CD8+ T cells and Tregs in their blood samples compared to UC patients and healthy dogs. Cytokine analysis demonstrated significant elevations in urinary cytokines (granulocyte-macrophage colony-stimulating factor, interferon-gamma [IFN-γ], interleukin (IL)-2, IL-6 IL-7, IL-8 and IL-15, IP-10, KC-like, IL-18, monocyte chemoattractant protein-1 and tumour necrosis factor-alpha). Several of these cytokines have been previously correlated with both lymphocyte-specific PD-1 expression (IFN-γ, IL-2, IL-7 and IL-15) in muscle-invasive urothelial carcinoma in humans. Our results provide evidence of urinary lymphocyte PD-1 expression and future studies could elucidate whether veterinary UC patients will respond favourably to anti-PD-1 immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Christopher J Pinard
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Anthony J Mutsaers
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.,Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - R Darren Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - J Paul Woods
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Samuel E Hocker
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.,Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
6
|
Ariafar A, Vahidi Y, Fakhimi M, Asadollahpour A, Erfani N, Faghih Z. Prognostic significance of CD4-positive regulatory T cells in tumor draining lymph nodes from patients with bladder cancer. Heliyon 2020; 6:e05556. [PMID: 33305045 PMCID: PMC7711140 DOI: 10.1016/j.heliyon.2020.e05556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction and methods To clarify the role of CD4+ regulatory T cells in bladder cancer, we investigated the frequency of these cells in tumor draining lymph nodes of 50 patients with bladder cancer who underwent radical cystectomy using flow cytometry method. We also assessed their association with prognosis and survival. Results On average, 30.13 ± 2.17% of lymphocytes in draining lymph nodes from patients with bladder cancer were positive for both CD4 and FOXP3 molecules. Analyses also showed that 9.92 ± 0.8% of CD4+ lymphocytes had a regulatory phenotype (CD4+CD25+FOXP3+CD127low/neg). The frequency of total CD4+FOXP3+ lymphocytes as well as regulatory T cells was significantly greater in patients with at least one tumor-involved lymph node compared to those with tumor-free nodes (P = 0.026 and P = 0.036, respectively). Mean FOXP3 expression in CD4+ lymphocytes was greater in patients with stage IV compared with those in stage III (P = 0.046). No other significant associations were found between the frequency of regulatory T cells and other clinicopathological characteristics or patient survival. Conclusions The increased frequency of regulatory T cells in patients with involved lymph nodes suggests that these cells may negatively regulate antitumor immune responses in draining lymph nodes. Our findings may have implications for immunotherapy-based treatments for bladder cancer.
Collapse
Affiliation(s)
- Ali Ariafar
- Urology-Oncology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Urology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasmin Vahidi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Fakhimi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ardalan Asadollahpour
- Department of Urology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrollah Erfani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Corresponding author.
| |
Collapse
|
7
|
Lillesand M, Kvikstad V, Mangrud OM, Gudlaugsson E, van Diermen-Hidle B, Skaland I, Baak JPA, Janssen EAM. Mitotic activity index and CD25+ lymphocytes predict risk of stage progression in non-muscle invasive bladder cancer. PLoS One 2020; 15:e0233676. [PMID: 32484812 PMCID: PMC7266352 DOI: 10.1371/journal.pone.0233676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/10/2020] [Indexed: 11/19/2022] Open
Abstract
In urothelial cell type non-muscle invasive urinary bladder carcinoma, TNM stage and WHO grade are widely used to classify patients into low and high‑risk groups for prognostic and therapeutic decision-making. However, stage and grade reproducibility and prediction accuracy are wanting. This may lead to suboptimal treatment. We evaluated whether proliferation features, nuclear area of the epithelial cancer cells and the composition of stromal and tumor infiltrating lymphocytes have independent prognostic value. In 183 primary non-muscle invasive bladder cancer patients with long follow-up (median for stage progression cohort: 119 months, range 5-173; median for tumor recurrence cohort: 82, range 3-165) proliferation features Ki67, PPH3 and Mitotic Activity Index (MAI), Mean Nuclear Area (MNA), lymphocyte subsets (CD8+, CD4+, CD25+) and plasma cells (CD138+) were assessed on consecutive sections. Post-resection instillation treatments (none, mitomycin, BCG) were strictly standardized during the intake period. Risk of recurrence was associated with expression of Ki67 (≤ 39 vs. > 39) and Multifocality (p = 0.01). Patients with low Ki67 had a higher recurrence rate than those with high Ki67. Lymphocyte composition did not predict recurrence. Stage progression was strongly associated with high values for MAI (>15) and CD25+ (>0.2%). In a multivariate analysis the combination of MAI and CD25+ was the single most prognostic feature (p<0.001). Validation of these results in additional, independent studies is warranted.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/immunology
- Carcinoma, Transitional Cell/pathology
- Disease Progression
- Disease-Free Survival
- Female
- Follow-Up Studies
- Humans
- Interleukin-2 Receptor alpha Subunit/metabolism
- Kaplan-Meier Estimate
- Ki-67 Antigen/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Middle Aged
- Mitotic Index
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/immunology
- Neoplasm Staging
- Prognosis
- Reproducibility of Results
- Urinary Bladder/pathology
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/immunology
- Urinary Bladder Neoplasms/mortality
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Melinda Lillesand
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- * E-mail:
| | - Vebjørn Kvikstad
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Mathematics and Natural Science, University of Stavanger, Stavanger, Norway
| | | | - Einar Gudlaugsson
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | | | - Ivar Skaland
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Jan P. A. Baak
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Jan Baak AS, Tananger, Norway
| | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Mathematics and Natural Science, University of Stavanger, Stavanger, Norway
| |
Collapse
|
8
|
The Tumor Microenvironment of Bladder Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1296:275-290. [PMID: 34185299 DOI: 10.1007/978-3-030-59038-3_17] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
|
9
|
Abstract
Bladder cancer is an important public health concern owing to its prevalence, high recurrence risk and treatment failures. Maintaining the equilibrium between prompt and effective immunity and an excessive and protracted immune response is critical for successful immune defence. This delicate balance is ensured by intrinsic or extrinsic immunoregulatory mechanisms. Intrinsic control of immune cell activation is mediated by stimulatory and inhibitory receptors expressed on the effector cell itself, whereas extrinsic control is mediated via other immune cells by cell-cell contact and/or secretion of inhibitory factors. Tumours can exacerbate these immunosuppressive pathways, fostering a tolerant microenvironment. These mechanisms have previously been poorly described in urothelial carcinoma, but a growing body of evidence highlights the key role of immune regulation in bladder cancer. This process includes immune checkpoints (mostly programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1)), as well as regulatory T cells, myeloid-derived suppressor cells, tumour-associated macrophages and type 2 innate and adaptive lymphocytes. For each component, quantitative and qualitative alterations, clinical relevance and potential targeting strategies are currently being explored. An improved understanding of immune regulation pathways in bladder cancer development, recurrence and progression will help in the design of novel diagnostic and prognostic tools as well as treatments.
Collapse
|
10
|
O’Donnell MA, Singh S, Sood R, Amlani J, Krishnamoorthy H, Shukla K, Mohanty N, Bhatia S, Chakraborty B, Desai N, Modi R, Shukla C, Vachhani K, Patel R, Kundu A, Khamar B. A Clinical Trial of the Intradermal TLR2 Agonist CADI-05 for BCG Recurrent and Unresponsive Non-Muscle Invasive Bladder Cancer. Bladder Cancer 2019. [DOI: 10.3233/blc-190211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - Sharwan Singh
- Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Rajeev Sood
- Dr. Ram Manohar Lohia Hospital & Post Graduate Institute of Medical Education and Research, New Delhi, India
| | | | | | | | | | | | | | - Nirav Desai
- Cadila Pharmaceuticals Limited, Ahmedabad, India
| | - Rajiv Modi
- Cadila Pharmaceuticals Limited, Ahmedabad, India
| | | | | | - Rashmi Patel
- Institute Of Kidney Disease and Research Centre, Ahmedabad, India
| | - Anup Kundu
- The Institute of Post-Graduate Medical Education and Research and Seth Sukhlal Karnani Memorial Hospital, Kolkata, India
| | | |
Collapse
|
11
|
Accurate Quantification of Residual Cancer Cells in Pelvic Washing Reveals Association with Cancer Recurrence Following Robot-Assisted Radical Cystectomy. J Urol 2019; 201:1105-1114. [PMID: 30730413 DOI: 10.1097/ju.0000000000000142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE Bladder cancer recurrence following cystectomy remains a significant cause of bladder cancer specific mortality. Residual cancer cells contribute to cancer recurrence due to tumor spillage or undetectable preexisting micrometastatic tumor clones. We detected and quantified residual cancer cells in pelvic washing using ultradeep targeted sequencing. We compared the levels of residual cancer cells with clinical variables and cancer recurrence. MATERIALS AND METHODS The primary tumor specimen was available in 17 patients who underwent robot-assisted radical cystectomy. All tumors had negative surgical margins. Pelvic washes and blood were collected intraoperatively before and after robot-assisted radical cystectomy, after pelvic lymph node dissection and in the suction fluid collected during the procedure. Two-step sequencing, including whole exome sequencing followed by ultradeep targeted sequencing (× greater than 50,000), was done to quantify residual cancer cells in each sample. Eight patients were excluded from study due to sample quality issues. The final analysis cohort comprised 9 patients. The residual cancer cell level was quantified for each sample as the relative cancer cell fraction and compared between time points. The peak relative cancer cell fraction of each patient was correlated with clinical and pathological variables. RESULTS Residual cancer cells were detected in approximately half of the pelvic washing specimens during or after but not before robot-assisted radical cystectomy. Higher residual cancer cell levels were associated with aggressive variant histology and cancer recurrence. Verifying the feasibility of using residual cancer cells as a novel biomarker for recurrence requires larger cohorts. CONCLUSIONS Detection of residual cancer cells in intraoperative peritoneal washes of patients with bladder cancer who undergo radical cystectomy may represent a robust biomarker of tumor aggressiveness and metastatic potential.
Collapse
|
12
|
Xiang M, Yuan W, Zhang W, Huang J. Expression of miR-490-5p, miR-148a-3p and miR-608 in bladder cancer and their effects on the biological characteristics of bladder cancer cells. Oncol Lett 2019; 17:4437-4442. [PMID: 30988813 PMCID: PMC6447928 DOI: 10.3892/ol.2019.10143] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Changes in the expression of miR-490-5p, miR-148a-3p and miR-608 in bladder cancer tissues were studied. A total of 30 patients with bladder cancer who had surgical resection in the Hunan Provincial People's Hospital (Changsha, China) from April 2015 to August 2016 were selected. RT-qPCR was used to detect the expression levels of miR-490-5p, miR-148a-3p and miR-608. The expression vectors of miR-490-5p, miR-148a-3p and miR-608 were respectively transfected and divided into three groups: blank cell group, gene transfection group (groups A-C) and negative transfection group (NC group). CCK8 was used to detect cell proliferation and flow cytometry was used to detect the condition of apoptosis of each group, and the Transwell chamber was used to detect the invasion ability of the cells. After the transfection, the expression level of miR-490-5p in group A was significantly higher than that in the NC and blank groups, and the expression level of miR-148a-3p in group B was significantly higher than that in the NC and blank groups. The expression level of miR-608 in group C was significantly higher than that in the NC and blank groups (P<0.001). The survival rates of the cells in groups A-C were significantly lower than those in the NC and blank groups at 48 and 72 h (P<0.001). After the transfection, the number of invasive cells and the apoptosis rates in groups A-C were significantly higher than those in the NC and blank groups (P<0.05). miR-490-5p, miR-148a-3p and miR-608 promoted proliferation of bladder cancer T24 cells and inhibited apoptosis of the cells and showed potential to become a new target for the future treatment of bladder cancer.
Collapse
Affiliation(s)
- Min Xiang
- Department of Urinary Surgery, Hunan Provincial People's Hospital, Changsha, Hunan 410005, P.R. China
| | - Wuxiong Yuan
- Department of Urinary Surgery, Hunan Provincial People's Hospital, Changsha, Hunan 410005, P.R. China
| | - Weiwei Zhang
- Department of Chinese Medicine, Hunan Provincial People's Hospital, Changsha, Hunan 410005, P.R. China
| | - Jie Huang
- Department of Emergency, Hunan Provincial People's Hospital, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
13
|
Chobrutskiy BI, Zaman S, Diviney A, Mihyu MM, Blanck G. T-cell receptor-α CDR3 domain chemical features correlate with survival rates in bladder cancer. J Cancer Res Clin Oncol 2018; 145:615-623. [DOI: 10.1007/s00432-018-2815-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023]
|
14
|
Magioncalda P, Martino M, Tardito S, Sterlini B, Conio B, Marozzi V, Adavastro G, Capobianco L, Russo D, Parodi A, Kalli F, Nasi G, Altosole T, Piaggio N, Northoff G, Fenoglio D, Inglese M, Filaci G, Amore M. White matter microstructure alterations correlate with terminally differentiated CD8+ effector T cell depletion in the peripheral blood in mania: Combined DTI and immunological investigation in the different phases of bipolar disorder. Brain Behav Immun 2018; 73:192-204. [PMID: 29723656 DOI: 10.1016/j.bbi.2018.04.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND White matter (WM) microstructural abnormalities and, independently, signs of immunological activation were consistently demonstrated in bipolar disorder (BD). However, the relationship between WM and immunological alterations as well as their occurrence in the various phases of BD remain unclear. METHOD In 60 type I BD patients - 20 in manic, 20 in depressive, 20 in euthymic phases - and 20 controls we investigated: (i) diffusion tensor imaging (DTI)-derived fractional anisotropy (FA), radial diffusivity (RD) and axial diffusivity (AD) using a tract-based spatial statistics (TBSS) approach; (ii) circulating T cell subpopulations frequencies, as well as plasma levels of different cytokines; (iii) potential relationships between WM and immunological data. RESULTS We found: (i) a significant widespread combined FA-RD alteration mainly in mania, with involvement of the body of corpus callosum (BCC) and superior corona radiata (SCR); (ii) significant increase in CD4+ T cells as well as significant decrease in CD8+ T cells and their subpopulations effector memory (CD8+ CD28-CD45RA-), terminal effector memory (CD8+ CD28-CD45RA+) and CD8+ IFNγ+ in mania; (iii) a significant relationship between WM and immunological alterations in the whole cohort, and a significant correlation of FA-RD abnormalities in the BCC and SCR with reduced frequencies of CD8+ terminal effector memory and CD8+ IFNγ+ T cells in mania only. CONCLUSIONS Our data show a combined occurrence of WM and immunological alterations in mania. WM abnormalities highly correlated with reduction in circulating CD8+ T cell subpopulations that are terminally differentiated effector cells prone to tissue migration, suggesting that these T cells could play a role in WM alteration in BD.
Collapse
Affiliation(s)
- Paola Magioncalda
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Matteo Martino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Samuele Tardito
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.
| | - Bruno Sterlini
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Benedetta Conio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Valentina Marozzi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Giulia Adavastro
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Laura Capobianco
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Daniel Russo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.
| | - Giorgia Nasi
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.
| | - Tiziana Altosole
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.
| | - Niccolò Piaggio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy.
| | - Georg Northoff
- University of Ottawa Brain and Mind Research Institute, and Mind Brain Imaging and Neuroethics Royal's Institute of Mental Health Research, University of Ottawa, Ottawa, Canada; Centre for Cognition and Brain Disorders, Hangzhou Normal University, Hangzhou, China; TMU Research Center for Brain and Consciousness, Taipei, Taiwan.
| | - Daniela Fenoglio
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Matilde Inglese
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy; Department of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, NY, USA.
| | - Gilberto Filaci
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
15
|
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. 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 2018; 13:e0204745. [PMID: 30261082 PMCID: PMC6160186 DOI: 10.1371/journal.pone.0204745] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 09/13/2018] [Indexed: 02/08/2023] Open
Abstract
Patients with a history of non-muscle-invasive bladder cancer sometimes have recurrence of tumors after transurethral resection of bladder tumor treatment. To find factors related to the recurrence of non-muscle-invasive bladder cancer, we examined tissue specimens taken at transurethral resection of bladder tumor as an initial treatment. We revealed the association between prognosis of non-muscle-invasive bladder cancer and infiltration of Foxp3+ T cells that suppress anti-tumor immunity in 115 primary non-muscle-invasive bladder cancer patients retrospectively identified and followed for at least 3 months after primary transurethral resection. In immunohistological staining, we counted the number of cells positive for CD3 and positive for CD3 and Foxp3 together and calculated the percentage of Foxp3+ T cells among the CD3+ T cells. The recurrence-free survival rate was calculated by the Kaplan-Meier method, and a Cox regression analysis of recurrence factors was performed. The median (interquartile range) percentage of Foxp3+ T cells in all cases was 17.1% (11.9, 11.4–23.3%). Compared by risk stratification, it was 11.4% (10.4, 7.8–18.2%) in the low-risk group (n = 32), 16.8% (12.6, 11.6–24.2%) in the intermediate-risk group (n = 45), and 22.0% (9.7, 16.4–26.1%) in the high-risk group (n = 38). The Kaplan-Meier survival analysis indicated that the Foxp3+ T cell high group (≥ 17.1%) had a worse RFS rate than did the low group (< 17.1%) (P = 0.006). In multivariate analysis, the percentage of Foxp3+ T cells was an independent risk factor for intravesical recurrence (hazard ratio 2.25). Thus, peritumoral Foxp3+ T cell infiltration was correlated to risk stratification and recurrence-free survival. Therefore, the percentage of Foxp3+ T cells in tumor specimens may predict a risk for intravesical recurrence.
Collapse
Affiliation(s)
- Ryosuke Murai
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
- * E-mail:
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Misako Nakayama
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hirohito Ishigaki
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kazuo Teramoto
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Mitsuhiro Narita
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tetsuya Yoshida
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Keiji Tomita
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Ken-ichi Kobayashi
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Akinori Wada
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masayuki Nagasawa
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shigehisa Kubota
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kazumasa Ogasawara
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
16
|
Pichler R, Fritz J, Zavadil C, Schäfer G, Culig Z, Brunner A. Tumor-infiltrating immune cell subpopulations influence the oncologic outcome after intravesical Bacillus Calmette-Guérin therapy in bladder cancer. Oncotarget 2018; 7:39916-39930. [PMID: 27221038 PMCID: PMC5129981 DOI: 10.18632/oncotarget.9537] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/05/2016] [Indexed: 12/22/2022] Open
Abstract
Although Bacillus Calmette-Guérin (BCG) is the most successful immunotherapy for high-risk non-muscle-invasive bladder cancer, approximately 30% of patients are unresponsive to treatment. New biomarkers are important to identify patients who will benefit most from BCG during a worldwide BCG shortage. Local immune cell subsets were measured on formalin-fixed, paraffin-embedded tissue sections of bladder cancer by immunohistochemistry, using monoclonal antibodies to tumor-associated macrophages (TAMs; CD68, CD163), B-lymphocytes (CD20) and T-lymphocyte subsets (CD3, CD4, CD8, GATA3, T-bet, FOXP3 and CD25). Cell densities in the lamina propria without invasion, at the invasive front if present, in the papillary tumor stroma, and in the neoplastic urothelium were calculated. Twenty-nine (72.5%) of 40 patients were classified as BCG responders after a mean follow-up of 35.3 months. A statistically significant association was observed for BCG failure with low density of CD4+ and GATA3+ T-cells, and increased expression of FOXP3+ and CD25+ regulatory T-cells (Tregs) as well as CD68+ and CD163+ TAMs. Survival analysis demonstrated prolonged recurrence-free survival (RFS) in patients with an increased count of CD4+ and GATA3+ T-cells. TAMs, Tregs and T-bet+ T-cells were inversely correlated with RFS. Thus, the tumor microenvironment seems to influence the therapeutic response to BCG, permitting an individualized treatment.
Collapse
Affiliation(s)
- Renate Pichler
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Josef Fritz
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Claudia Zavadil
- Department of Pathology, Division of General Pathology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Georg Schäfer
- Department of Pathology, Division of General Pathology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Zoran Culig
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Andrea Brunner
- Department of Pathology, Division of General Pathology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
17
|
Fenoglio D, Dentone C, Signori A, Di Biagio A, Parodi A, Kalli F, Nasi G, Curto M, Cenderello G, De Leo P, Bartolacci V, Orofino G, Nicolini LA, Taramasso L, Fiorillo E, Orrù V, Traverso P, Bruzzone B, Ivaldi F, Mantia E, Guerra M, Negrini S, Giacomini M, Bhagani S, Filaci G. CD8 +CD28 -CD127 loCD39 + regulatory T-cell expansion: A new possible pathogenic mechanism for HIV infection? J Allergy Clin Immunol 2017; 141:2220-2233.e4. [PMID: 29103633 DOI: 10.1016/j.jaci.2017.08.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/10/2017] [Accepted: 08/28/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND HIV-associated immunodeficiency is related to loss of CD4+ T cells. This mechanism does not explain certain manifestations of HIV disease, such as immunodeficiency events in patients with greater than 500 CD4+ T cells/μL. CD8+CD28-CD127loCD39+ T cells are regulatory T (Treg) lymphocytes that are highly concentrated within the tumor microenvironment and never analyzed in the circulation of HIV-infected patients. OBJECTIVES We sought to analyze the frequency of CD8+CD28-CD127loCD39+ Treg cells in the circulation of HIV-infected patients. METHODS The frequency of circulating CD8+CD28-CD127loCD39+ Treg cells was analyzed and correlated with viral load and CD4+ T-cell counts/percentages in 93 HIV-1-infected patients subdivided as follows: naive (n = 63), elite controllers (n = 19), long-term nonprogressors (n = 7), and HIV-infected patients affected by tumor (n = 4). The same analyses were performed in HIV-negative patients with cancer (n = 53), hepatitis C virus-infected patients (n = 17), and healthy donors (n = 173). RESULTS HIV-infected patients had increased circulating levels of functional CD8+CD28-CD127loCD39+ Treg cells. These cells showed antigen specificity against HIV proteins. Their frequency after antiretroviral therapy (ART) correlated with HIV viremia, CD4+ T-cell counts, and immune activation markers, suggesting their pathogenic involvement in AIDS- or non-AIDS-related complications. Their increase after initiation of ART heralded a lack of virologic or clinical response, and hence their monitoring is clinically relevant. CONCLUSION HIV infection induces remarkable expansion of CD8+CD28-CD127loCD39+ Treg cells, the frequency of which correlates with both clinical disease and signs of chronic immune cell activation. Monitoring their frequency in the circulation is a new marker of response to ART when effects on viremia and clinical response are not met.
Collapse
Affiliation(s)
- Daniela Fenoglio
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Chiara Dentone
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Infectious Diseases Department, Sanremo Hospital, Imperia, Italy
| | - Alessio Signori
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Antonio Di Biagio
- Infectious Disease Unit, IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Giorgia Nasi
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Monica Curto
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | | | | | | - Giancarlo Orofino
- Infectious Diseases Department, Amedeo di Savoia Hospital, Turin, Italy
| | - Laura Ambra Nicolini
- Department of Health Sciences, University of Genoa, Genoa, Italy; Infectious Disease Unit, IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Lucia Taramasso
- Infectious Disease Unit, IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Sede Secondaria IRGB, Lanusei, Italy
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Sede Secondaria IRGB, Lanusei, Italy
| | - Paolo Traverso
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy; Department of Surgical Science and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Bianca Bruzzone
- Hygiene Unit, Infectious Disease Unit, IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Federico Ivaldi
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Eugenio Mantia
- Infectious Diseases Department, SS Antonio, Biagio, Cesare Arrigo Hospital, Alessandria, Italy
| | - Michele Guerra
- Infectious Diseases Department, Sant'Andrea Hospital, La Spezia, Italy
| | - Simone Negrini
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Mauro Giacomini
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Informatics, Bioengineering, Robotic and System Engineering, University of Genoa, Genoa, Italy
| | - Sanjay Bhagani
- Department of Infectious Diseases/HIV Medicine, Royal Free Hospital, National Health Service, London, United Kingdom
| | - Gilberto Filaci
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Azienda Ospedaliero Universitaria San Martino, IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy.
| |
Collapse
|
18
|
Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, Christie M, van de Vijver K, Estrada MV, Gonzalez-Ericsson PI, Sanders M, Solomon B, Solinas C, Van den Eynden GGGM, Allory Y, Preusser M, Hainfellner J, Pruneri G, Vingiani A, Demaria S, Symmans F, Nuciforo P, Comerma L, Thompson EA, Lakhani S, Kim SR, Schnitt S, Colpaert C, Sotiriou C, Scherer SJ, Ignatiadis M, Badve S, Pierce RH, Viale G, Sirtaine N, Penault-Llorca F, Sugie T, Fineberg S, Paik S, Srinivasan A, Richardson A, Wang Y, Chmielik E, Brock J, Johnson DB, Balko J, Wienert S, Bossuyt V, Michiels S, Ternes N, Burchardi N, Luen SJ, Savas P, Klauschen F, Watson PH, Nelson BH, Criscitiello C, O’Toole S, Larsimont D, de Wind R, Curigliano G, André F, Lacroix-Triki M, van de Vijver M, Rojo F, Floris G, Bedri S, Sparano J, Rimm D, Nielsen T, Kos Z, Hewitt S, Singh B, Farshid G, Loibl S, Allison KH, Tung N, Adams S, Willard-Gallo K, Horlings HM, Gandhi L, Moreira A, Hirsch F, Dieci MV, Urbanowicz M, Brcic I, Korski K, Gaire F, Koeppen H, Lo A, Giltnane J, Ziai J, Rebelatto MC, Steele KE, Zha J, Emancipator K, Juco JW, Denkert C, Reis-Filho J, Loi S, Fox SB. Assessing Tumor-Infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in Melanoma, Gastrointestinal Tract Carcinomas, Non-Small Cell Lung Carcinoma and Mesothelioma, Endometrial and Ovarian Carcinomas, Squamous Cell Carcinoma of the Head and Neck, Genitourinary Carcinomas, and Primary Brain Tumors. Adv Anat Pathol 2017; 24:311-335. [PMID: 28777143 PMCID: PMC5638696 DOI: 10.1097/pap.0000000000000161] [Citation(s) in RCA: 481] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Assessment of the immune response to tumors is growing in importance as the prognostic implications of this response are increasingly recognized, and as immunotherapies are evaluated and implemented in different tumor types. However, many different approaches can be used to assess and describe the immune response, which limits efforts at implementation as a routine clinical biomarker. In part 1 of this review, we have proposed a standardized methodology to assess tumor-infiltrating lymphocytes (TILs) in solid tumors, based on the International Immuno-Oncology Biomarkers Working Group guidelines for invasive breast carcinoma. In part 2 of this review, we discuss the available evidence for the prognostic and predictive value of TILs in common solid tumors, including carcinomas of the lung, gastrointestinal tract, genitourinary system, gynecologic system, and head and neck, as well as primary brain tumors, mesothelioma and melanoma. The particularities and different emphases in TIL assessment in different tumor types are discussed. The standardized methodology we propose can be adapted to different tumor types and may be used as a standard against which other approaches can be compared. Standardization of TIL assessment will help clinicians, researchers and pathologists to conclusively evaluate the utility of this simple biomarker in the current era of immunotherapy.
Collapse
Affiliation(s)
- Shona Hendry
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory/Breast International Group, Institut Jules Bordet, Brussels, Belgium
- Department of Pathology and TCRU, GZA, Antwerp, Belgium
| | - Thomas Gevaert
- Department of Development and Regeneration, Laboratory of Experimental Urology, KU Leuven, Leuven, Belgium
- Department of Pathology, AZ Klina, Brasschaat, Belgium
| | - Prudence A. Russell
- Department of Anatomical Pathology, St Vincent’s Hospital Melbourne, Fitzroy, Australia
- Department of Pathology, University of Melbourne, Parkville, Australia
| | - Tom John
- Department of Medical Oncology, Austin Health, Heidelberg, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | - Bibhusal Thapa
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
| | - Michael Christie
- Department of Anatomical Pathology, Royal Melbourne Hospital, Parkville, Australia
| | - Koen van de Vijver
- Divisions of Diagnostic Oncology & Molecular Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - M. Valeria Estrada
- Department of Pathology, School of Medicine, University of California, San Diego, USA
| | | | - Melinda Sanders
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| | - Benjamin Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gert GGM Van den Eynden
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
- Department of Pathology, GZA Ziekenhuizen, Antwerp, Belgium
| | - Yves Allory
- Université Paris-Est, Créteil, France
- INSERM, UMR 955, Créteil, France
- Département de pathologie, APHP, Hôpital Henri-Mondor, Créteil, France
| | - Matthias Preusser
- Department of Medicine, Clinical Division of Oncology, Comprehensive Cancer Centre Vienna, Medical University of Vienna, Vienna, Austria
| | - Johannes Hainfellner
- Institute of Neurology, Comprehensive Cancer Centre Vienna, Medical University of Vienna, Vienna, Austria
| | - Giancarlo Pruneri
- European Institute of Oncology, Milan, Italy
- University of Milan, School of Medicine, Milan, Italy
| | - Andrea Vingiani
- European Institute of Oncology, Milan, Italy
- University of Milan, School of Medicine, Milan, Italy
| | - Sandra Demaria
- New York University Medical School, New York, USA
- Perlmutter Cancer Center, New York, USA
| | - Fraser Symmans
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
| | - Laura Comerma
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, Barcelona, Spain
| | | | - Sunil Lakhani
- Centre for Clinical Research and School of Medicine, The University of Queensland, Brisbane, Australia
- Pathology Queensland, Royal Brisbane and Women’s Hospital, Brisbane, Australia
| | - Seong-Rim Kim
- National Surgical Adjuvant Breast and Bowel Project Operations Center/NRG Oncology, Pittsburgh, Pennsylvania
| | - Stuart Schnitt
- Cancer Research Institute and Department of Pathology, Beth Israel Deaconess Cancer Center, Boston, USA
- Harvard Medical School, Boston, USA
| | - Cecile Colpaert
- Department of Pathology, GZA Ziekenhuizen, Sint-Augustinus, Wilrijk, Belgium
| | - Christos Sotiriou
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Stefan J. Scherer
- Academic Medical Innovation, Novartis Pharmaceuticals Corporation, East Hanover, USA
| | - Michail Ignatiadis
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, USA
| | - Robert H. Pierce
- Cancer Immunotherapy Trials Network, Central Laboratory and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Giuseppe Viale
- Department of Pathology, Istituto Europeo di Oncologia, University of Milan, Milan, Italy
| | - Nicolas Sirtaine
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Frederique Penault-Llorca
- Department of Surgical Pathology and Biopathology, Jean Perrin Comprehensive Cancer Centre, Clermont-Ferrand, France
- University of Auvergne UMR1240, Clermont-Ferrand, France
| | - Tomohagu Sugie
- Department of Surgery, Kansai Medical School, Hirakata, Japan
| | - Susan Fineberg
- Montefiore Medical Center, Bronx, New York, USA
- The Albert Einstein College of Medicine, Bronx, New York, USA
| | - Soonmyung Paik
- National Surgical Adjuvant Breast and Bowel Project Operations Center/NRG Oncology, Pittsburgh, Pennsylvania
- Severance Biomedical Science Institute and Department of Medical Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Ashok Srinivasan
- National Surgical Adjuvant Breast and Bowel Project Operations Center/NRG Oncology, Pittsburgh, Pennsylvania
| | - Andrea Richardson
- Harvard Medical School, Boston, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Providence, USA
- Warren Alpert Medical School of Brown University, Providence, USA
| | - Ewa Chmielik
- Tumor Pathology Department, Maria Sklodowska-Curie Memorial Cancer Center, Gliwice, Poland
- Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Jane Brock
- Harvard Medical School, Boston, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, USA
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, USA
- Vanderbilt Ingram Cancer Center, Nashville, USA
| | - Justin Balko
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, USA
- Vanderbilt Ingram Cancer Center, Nashville, USA
| | - Stephan Wienert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- VMscope GmbH, Berlin, Germany
| | - Veerle Bossuyt
- Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - Stefan Michiels
- Service de Biostatistique et d’Epidémiologie, Gustave Roussy, CESP, Inserm U1018, Université-Paris Sud, Université Paris-Saclay, Villejuif, France
| | - Nils Ternes
- Service de Biostatistique et d’Epidémiologie, Gustave Roussy, CESP, Inserm U1018, Université-Paris Sud, Université Paris-Saclay, Villejuif, France
| | | | - Stephen J. Luen
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Peter Savas
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Peter H. Watson
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Brad H. Nelson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Sandra O’Toole
- The Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Australia
- Australian Clinical Labs, Bella Vista, Australia
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Roland de Wind
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Fabrice André
- INSERM Unit U981, and Department of Medical Oncology, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université Paris Sud, Kremlin-Bicêtre, France
| | - Magali Lacroix-Triki
- INSERM Unit U981, and Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Mark van de Vijver
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Federico Rojo
- Pathology Department, IIS-Fundacion Jimenez Diaz, UAM, Madrid, Spain
| | - Giuseppe Floris
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Shahinaz Bedri
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Joseph Sparano
- Department of Oncology, Montefiore Medical Centre, Albert Einstein College of Medicine, Bronx, USA
| | - David Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - Torsten Nielsen
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Zuzana Kos
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Canada
| | - Stephen Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Baljit Singh
- Department of Pathology, New York University Langone Medical Centre, New York, USA
| | - Gelareh Farshid
- Directorate of Surgical Pathology, SA Pathology, Adelaide, Australia
- Discipline of Medicine, Adelaide University, Adelaide, Australia
| | | | | | - Nadine Tung
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, USA
| | - Sylvia Adams
- New York University Medical School, New York, USA
- Perlmutter Cancer Center, New York, USA
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Hugo M. Horlings
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Leena Gandhi
- Perlmutter Cancer Center, New York, USA
- Dana-Farber Cancer Institute, Boston, USA
| | - Andre Moreira
- Pulmonary Pathology, New York University Center for Biospecimen Research and Development, New York University, New York, USA
| | - Fred Hirsch
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Maria Urbanowicz
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - Iva Brcic
- Institute of Pathology, Medical University of Graz, Austria
| | - Konstanty Korski
- Pathology and Tissue Analytics, Roche Innovation Centre Munich, Penzberg, Germany
| | - Fabien Gaire
- Pathology and Tissue Analytics, Roche Innovation Centre Munich, Penzberg, Germany
| | - Hartmut Koeppen
- Research Pathology, Genentech Inc., South San Francisco, USA
| | - Amy Lo
- Research Pathology, Genentech Inc., South San Francisco, USA
- Department of Pathology, Stanford University, Palo Alto, USA
| | | | - James Ziai
- Research Pathology, Genentech Inc., South San Francisco, USA
| | | | | | - Jiping Zha
- Translational Sciences, MedImmune, Gaithersberg, USA
| | | | | | - Carsten Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jorge Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Sherene Loi
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Stephen B. Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
19
|
Solinas C, Chanzá NM, Awada A, Scartozzi M. The immune infiltrate in prostate, bladder and testicular tumors: An old friend for new challenges. Cancer Treat Rev 2016; 53:138-145. [PMID: 28113097 DOI: 10.1016/j.ctrv.2016.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 12/16/2016] [Accepted: 12/18/2016] [Indexed: 12/20/2022]
Abstract
In genito-urinary tumors immunotherapy has been administered for a long time: Calmette-Guèrin Bacillus as adjuvant treatment in high risk patients with non muscle invasive urothelial bladder cancer and interleukin-2 and interferon-α in metastatic kidney cancer. The vaccine Sipuleucel-T has been approved by United States Food and Drug Administration for the treatment of castration resistant prostate cancer patients with asymptomatic or minimally symptomatic disease, given the 22% reduction of mortality risk in this group. Recently immunotherapeutic agents targeting inhibitory immune checkpoint molecules lead to improved outcomes and lasting anti-tumor effects in a variety of hematological and solid malignancies, including urogenital tumors. The benefit from these treatments has been observed only in a proportion of subjects, raising a need in optimizing patients' selection for immune checkpoint blockade. The composition and activity of a pre-existing immune infiltrate may aid in identifying ideal candidates to immunotherapy, with possible implications for the clinical management of neoplastic diseases from earlier to later stages.
Collapse
Affiliation(s)
- Cinzia Solinas
- Molecular Immunology Unit, Université Libre de Bruxelles, Institut Jules Bordet, Brussels, Belgium.
| | - Nieves Martinez Chanzá
- Medical Oncology, Université Libre de Bruxelles, Institut Jules Bordet, Brussels, Belgium; Medical Oncology, Université Libre de Bruxelles, Erasme Hospital, Brussels, Belgium.
| | - Ahmad Awada
- Medical Oncology, Université Libre de Bruxelles, Institut Jules Bordet, Brussels, Belgium.
| | | |
Collapse
|
20
|
Yan X, Wu C, Chen T, Santos MM, Liu CL, Yang C, Zhang L, Ren J, Liao S, Guo H, Sukhova GK, Shi GP. Cathepsin S inhibition changes regulatory T-cell activity in regulating bladder cancer and immune cell proliferation and apoptosis. Mol Immunol 2016; 82:66-74. [PMID: 28033540 DOI: 10.1016/j.molimm.2016.12.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 12/29/2022]
Abstract
Regulatory T cells (Tregs) are immune suppressive cells, but their roles in tumor growth have been elusive, depending on tumor type or site. Our prior study demonstrated a role of cathepsin S (CatS) in reducing Treg immunosuppressive activity. Therefore, CatS inhibition in Tregs may exacerbate tumor growth. Using mouse bladder carcinoma MB49 cell subcutaneous implant tumor model, we detected no difference in tumor growth, whether mice were given saline- or CatS inhibitor-treated Tregs. However, mice that received inhibitor-treated Tregs had fewer splenic and tumor Tregs, and lower levels of tumor and splenic cell proliferation than mice that received saline-treated Tregs. In vitro, inhibitor-treated Tregs showed lower proliferation and higher apoptosis than saline-treated Tregs when cells were exposed to MB49. In contrast, both types of Tregs showed no difference in proliferation when they were co-cultured with normal splenocytes. Inhibitor-treated Tregs had less apoptosis in splenocytes, but more apoptosis in splenocytes with MB49 conditioned media than saline-treated Tregs. In turn, we detected less proliferation and more apoptosis of MB94 cells after co-culture with inhibitor-treated Tregs, compared with saline-treated Tregs. B220+ B-cell, CD4+ T-cell, and CD8+ T-cell proliferation and apoptosis were also lower in splenocytes co-cultured with inhibitor-treated Tregs than with saline-treated Tregs. Under the same conditions, the addition of cancer cell-conditioned media greatly increased CD8+ T-cell proliferation and reduced CD8+ T-cell apoptosis. These observations suggest that CatS inhibition of Tregs may reduce overall T-cell immunity under normal conditions, but enhance CD8+ T-cell immunity in the presence of cancer cells.
Collapse
Affiliation(s)
- Xiang Yan
- Department of Urology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Chun Wu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Cardiology, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and technology, Wuhan, 430022, China
| | - Tao Chen
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Marcela M Santos
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Cong-Lin Liu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Chongzhe Yang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lijun Zhang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jingyuan Ren
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sha Liao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hongqiang Guo
- Department of Urology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China
| | - Galina K Sukhova
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Bahria-Sediki IB, Yousfi N, Paul C, Chebil M, Cherif M, Zermani R, El Gaaied ABA, Bettaieb A. Clinical significance of T-bet, GATA-3, and Bcl-6 transcription factor expression in bladder carcinoma. J Transl Med 2016; 14:144. [PMID: 27237631 PMCID: PMC4885121 DOI: 10.1186/s12967-016-0891-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/02/2016] [Indexed: 11/15/2022] Open
Abstract
Background The aim of this study was to investigate the clinical significance of three immune cell-related transcription factors, T-bet, GATA-3 and Bcl-6 in bladder cancer in Tunisian patients. Methods Expression of T-bet, GATA-3 and Bcl-6 genes was assessed using RT-qPCR in 65 bladder cancers from patients: 32 being diagnosed as low- and medium-grade, 31 as high-grade, 25 as muscle invasive stage and 39 as non-muscle invasive stage. Gene expression was statistically correlated according to the grade, the stage, tobacco consumption, the BCG response and disease severity. Results T-bet levels in patients with high-grade bladder cancer were significantly elevated compared to patients with low- or medium-grade bladder cancer (p = 0.005). In invasive carcinoma (T2–T4), the T-bet levels were significantly higher than in superficial non-invasive bladder tumors (Tis, Ta, and T1) (p = 0.02). However, T-bet is predictive of the response to BCG. Its expression is high in good responders to BCG (p = 0.02). In contrast, the expression of GATA-3 and Bcl-6 in non-invasive carcinoma (p = 0.008 and p = 0.0003) and in patients with low- and medium-grade cancers (p = 0.001 and p < 0.0001) is significantly higher than in invasive bladder tumors and in patients with high-grade bladder carcinoma, respectively. In addition, heavy smokers, whose tumors express low levels of GATA-3 and Bcl-6, are poor responders to BCG (p = 0.01 and p = 0.03). Finally, better patient survival correlated with GATA-3 (p = 0.04) and Bcl-6 (p = 0.04) but not T-bet expression. Conclusions Our results suggest that T-bet expression in bladder tumors could be a positive prognostic indicator of BCG therapy, even if high levels are found in high-grade and stage of the disease. However, GATA-3 and Bcl-6 expression could be considered as predictive factors for good patient survival.
Collapse
Affiliation(s)
- Islem Ben Bahria-Sediki
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EPHE, PSL Research University, 75014, Paris, France.,Université Bourgogne Franche-Comté, EA7269, 21000, Dijon, France.,Laboratoire de Génétique, Immunologie et Pathologies Humaines, Faculté de Sciences Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Nadhir Yousfi
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EPHE, PSL Research University, 75014, Paris, France.,Université Bourgogne Franche-Comté, EA7269, 21000, Dijon, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EPHE, PSL Research University, 75014, Paris, France.,Université Bourgogne Franche-Comté, EA7269, 21000, Dijon, France
| | | | | | | | - Amel Ben Ammar El Gaaied
- Laboratoire de Génétique, Immunologie et Pathologies Humaines, Faculté de Sciences Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EPHE, PSL Research University, 75014, Paris, France. .,Université Bourgogne Franche-Comté, EA7269, 21000, Dijon, France. .,UFR des Sciences de Santé, 7 boulevard Jeanne d'Arc, BP 87900, 21079, Dijon, France.
| |
Collapse
|