Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 18(8): 940-950, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28628092

ABSTRACT

Therapies that boost the anti-tumor responses of cytotoxic T lymphocytes (CTLs) have shown promise; however, clinical responses to the immunotherapeutic agents currently available vary considerably, and the molecular basis of this is unclear. We performed transcriptomic profiling of tumor-infiltrating CTLs from treatment-naive patients with lung cancer to define the molecular features associated with the robustness of anti-tumor immune responses. We observed considerable heterogeneity in the expression of molecules associated with activation of the T cell antigen receptor (TCR) and of immunological-checkpoint molecules such as 4-1BB, PD-1 and TIM-3. Tumors with a high density of CTLs showed enrichment for transcripts linked to tissue-resident memory cells (TRM cells), such as CD103, and CTLs from CD103hi tumors displayed features of enhanced cytotoxicity. A greater density of TRM cells in tumors was predictive of a better survival outcome in lung cancer, and this effect was independent of that conferred by CTL density. Here we define the 'molecular fingerprint' of tumor-infiltrating CTLs and identify potentially new targets for immunotherapy.


Subject(s)
Adenocarcinoma/immunology , Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Immunologic Memory/immunology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Carcinoma, Squamous Cell/mortality , Female , Gene Expression Profiling , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Immunotherapy , Integrin alpha Chains/genetics , Lung Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/genetics , Receptors, Antigen, T-Cell/genetics , Squamous Cell Carcinoma of Head and Neck , Survival Rate , T-Lymphocytes, Cytotoxic/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
2.
Nature ; 605(7911): 741-746, 2022 05.
Article in English | MEDLINE | ID: mdl-35508656

ABSTRACT

Phosphoinositide 3-kinase δ (PI3Kδ) has a key role in lymphocytes, and inhibitors that target this PI3K have been approved for treatment of B cell malignancies1-3. Although studies in mouse models of solid tumours have demonstrated that PI3Kδ inhibitors (PI3Kδi) can induce anti-tumour immunity4,5, its effect on solid tumours in humans remains unclear. Here we assessed the effects of the PI3Kδi AMG319 in human patients with head and neck cancer in a neoadjuvant, double-blind, placebo-controlled randomized phase II trial (EudraCT no. 2014-004388-20). PI3Kδ inhibition decreased the number of tumour-infiltrating regulatory T (Treg) cells and enhanced the cytotoxic potential of tumour-infiltrating T cells. At the tested doses of AMG319, immune-related adverse events (irAEs) required treatment to be discontinued in 12 out of 21 of patients treated with AMG319, suggestive of systemic effects on Treg cells. Accordingly, in mouse models, PI3Kδi decreased the number of Treg cells systemically and caused colitis. Single-cell RNA-sequencing analysis revealed a PI3Kδi-driven loss of tissue-resident colonic ST2 Treg cells, accompanied by expansion of pathogenic T helper 17 (TH17) and type 17 CD8+ T (TC17) cells, which probably contributed to toxicity; this points towards a specific mode of action for the emergence of irAEs. A modified treatment regimen with intermittent dosing of PI3Kδi in mouse models led to a significant decrease in tumour growth without inducing pathogenic T cells in colonic tissue, indicating that alternative dosing regimens might limit toxicity.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Adenosine/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Head and Neck Neoplasms/drug therapy , Humans , Immunotherapy , Mice , Phosphatidylinositol 3-Kinases , Quinolines/therapeutic use , T-Lymphocytes, Regulatory
3.
Neuroendocrinology ; 112(4): 370-383, 2022.
Article in English | MEDLINE | ID: mdl-34157710

ABSTRACT

INTRODUCTION: Neuroendocrine tumours (NETs) are rare tumours with an increasing incidence. While low- and intermediate-grade pancreatic NET (PanNET) and small intestinal NET (siNET) are slow growing, they have a relatively high rate of metastasizing to the liver, leading to substantially worse outcomes. In many solid tumours, the outcome is determined by the quality of the antitumour immune response. However, the quality and significance of antitumour responses in NETs are incompletely understood. This study provides clinico-pathological analyses of the tumour immune microenvironment in PanNET and siNETs. METHODS: Formalin-fixed paraffin-embedded tissue from consecutive resected PanNETs (61) and siNETs (131) was used to construct tissue microarrays (TMAs); 1-mm cores were taken from the tumour centre, stroma, tumour edge, and adjacent healthy tissue. TMAs were stained with antibodies against CD8, CD4, CD68, FoxP3, CD20, and NCR1. T-cell counts were compared with counts from lung cancers. RESULTS: For PanNET, median counts were CD8+ 35.4 cells/mm2, CD4+ 7.6 cells/mm2, and CD68+ macrophages 117.7 cells/mm2. For siNET, there were CD8+ 39.2 cells/mm2, CD4+ 24.1 cells/mm2, and CD68+ 139.2 cells/mm2. The CD8+ cell density in the tumour and liver metastases were significantly lower than in the adjacent normal tissues, without evidence of a cell-rich area at the tumour edge that might have suggested immune exclusion. T-cell counts in lung cancer were significantly higher than those in PanNET and siNETs: CD8+ 541 cells/mm2 and CD4+ 861 cells/mm2 (p ≤ 0.0001). CONCLUSION: PanNETs and siNETs are immune cold with no evidence of T cell exclusion; the low density of immune infiltrates indicates poor antitumour immune responses.


Subject(s)
Neuroendocrine Tumors , Pancreatic Neoplasms , Stomach Neoplasms , Humans , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Prognosis , Tumor Microenvironment
5.
Br J Cancer ; 120(3): 356-367, 2019 02.
Article in English | MEDLINE | ID: mdl-30655616

ABSTRACT

BACKGROUND: Metabolic changes in tumour cells are used in clinical imaging and may provide potential therapeutic targets. Human papillomavirus (HPV) status is important in classifying head and neck cancers (HNSCC), identifying a distinct clinical phenotype; metabolic differences between these HNSCC subtypes remain poorly understood. METHODS: We used RNA sequencing to classify the metabolic expression profiles of HPV+ve and HPV-ve HNSCC, performed a meta-analysis on FDG-PET imaging characteristics and correlated results with in vitro extracellular flux analysis of HPV-ve and HPV+ve HNSCC cell lines. The monocarboxylic acid transporter-1 (MCT1) was identified as a potential metabolic target and tested in functional assays. RESULTS: Specific metabolic profiles were associated with HPV status, not limited to carbohydrate metabolism. There was dominance of all energy pathways in HPV-negative disease, with elevated expression of genes associated with glycolysis and oxidative phosphorylation. In vitro analysis confirmed comparative increased rates of oxidative phosphorylation and glycolysis in HPV-negative cell lines. PET SUV(max) scores however were unable to reliably differentiate between HPV-positive and HPV-negative tumours. MCT1 expression was significantly increased in HPV-negative tumours, and inhibition suppressed tumour cell invasion, colony formation and promoted radiosensitivity. CONCLUSION: HPV-positive and negative HNSCC have different metabolic profiles which may have potential therapeutic applications.


Subject(s)
Monocarboxylic Acid Transporters/genetics , Papillomaviridae/pathogenicity , Papillomavirus Infections/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Symporters/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Glycolysis/genetics , Humans , Monocarboxylic Acid Transporters/isolation & purification , Monocarboxylic Acid Transporters/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oxidative Phosphorylation , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus Infections/diagnostic imaging , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Positron-Emission Tomography , Radiation Tolerance , Sequence Analysis, RNA , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/virology , Symporters/isolation & purification , Symporters/metabolism
6.
Dermatol Ther ; 32(3): e12800, 2019 05.
Article in English | MEDLINE | ID: mdl-30523664

ABSTRACT

Cyclooxygenase-2 (COX-2) and its metabolic product prostaglandin E2 (PGE2 ) are induced in response to growth factors, inflammatory cytokines, tumor promoters, activated oncogenes, and, in the skin, ultraviolet (UV) radiation. Accumulating evidence suggests a role for the COX-2/PGE2 pathway in tumorigenesis in various tissue types including cutaneous squamous cell carcinoma. There is also strong evidence for a role in the development of actinic keratoses (AKs) - common dysplastic lesions of the skin associated with UV radiation overexposure - considered as part of a continuum with skin cancer. Non-steroidal anti-inflammatory drugs (NSAIDs) exert their anti-inflammatory, analgesic, and antipyretic effects by reversibly or irreversibly acetylating COX isoforms, inhibiting downstream prostaglandins, and may have a chemopreventive role in malignancies, including skin cancer. Topical treatment of AK lesions with the NSAID diclofenac sodium 3% in combination with hyaluronic acid 2.5% has been shown to be effective and well tolerated, although the mechanism of action remains to be elucidated.


Subject(s)
Diclofenac/administration & dosage , Hyaluronic Acid/administration & dosage , Keratosis, Actinic/drug therapy , Administration, Cutaneous , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carcinoma, Squamous Cell/prevention & control , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/pharmacology , Diclofenac/pharmacology , Humans , Keratosis, Actinic/pathology , Skin Neoplasms/prevention & control
7.
Carcinogenesis ; 39(6): 798-807, 2018 05 28.
Article in English | MEDLINE | ID: mdl-29506142

ABSTRACT

The dissemination of cancer cells to local and distant sites depends on a complex and poorly understood interplay between malignant cells and the cellular and non-cellular components surrounding them, collectively termed the tumour microenvironment. One of the most abundant cell types of the tumour microenvironment is the fibroblast, which becomes corrupted by locally derived cues such as TGF-ß1 and acquires an altered, heterogeneous phenotype (cancer-associated fibroblasts, CAF) supportive of tumour cell invasion and metastasis. Efforts to develop new treatments targeting the tumour mesenchyme are hampered by a poor understanding of the mechanisms underlying the development of CAF. Here, we examine the contribution of microRNA to the development of experimentally-derived CAF and correlate this with changes observed in CAF derived from tumours. Exposure of primary normal human fibroblasts to TGF-ß1 resulted in the acquisition of a myofibroblastic CAF-like phenotype. This was associated with increased expression of miR-145, a miRNA predicted in silico to target multiple components of the TGF-ß signalling pathway. miR-145 was also overexpressed in CAF derived from oral cancers. Overexpression of miR-145 blocked TGF-ß1-induced myofibroblastic differentiation and reverted CAF towards a normal fibroblast phenotype. We conclude that miR-145 is a key regulator of the CAF phenotype, acting in a negative feedback loop to dampen acquisition of myofibroblastic traits, a key feature of CAF associated with poor disease outcome.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , MicroRNAs/metabolism , Mouth Neoplasms/metabolism , Transforming Growth Factor beta1/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Humans , Myofibroblasts/metabolism , Phenotype , Signal Transduction/physiology , Tumor Microenvironment/physiology
8.
Br J Cancer ; 118(9): 1200-1207, 2018 05.
Article in English | MEDLINE | ID: mdl-29593339

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) form the major stromal component of the tumour microenvironment (TME). The present study aimed to examine the proteomic profiles of CAFs vs. normal fibroblasts (NOFs) from patients with oesophageal adenocarcinoma to gain insight into their pro-oncogenic phenotype. METHODS: CAFs/NOFs from four patients were sub-cultured and analysed using quantitative proteomics. Differentially expressed proteins (DEPs) were subjected to bioinformatics and compared with published proteomics and transcriptomics  datasets. RESULTS: Principal component analysis of all profiled proteins showed that CAFs had high heterogeneity and clustered separately from NOFs. Bioinformatics interrogation of the DEPs demonstrated inhibition of adhesion of epithelial cells, adhesion of connective tissue cells and cell death of fibroblast cell lines in CAFs vs. NOFs (p < 0.0001). KEGG pathway analysis showed a significant enrichment of the insulin-signalling pathway (p = 0.03). Gene ontology terms related with myofibroblast phenotype, metabolism, cell adhesion/migration, hypoxia/oxidative stress, angiogenesis, immune/inflammatory response were enriched in CAFs vs. NOFs. Nestin, a stem-cell marker up-regulated in CAFs vs. NOFs, was confirmed to be expressed in the TME with immunohistochemistry. CONCLUSIONS: The identified pathways and participating proteins may provide novel insight on the tumour-promoting properties of CAFs and unravel novel adjuvant therapeutic targets in the TME.


Subject(s)
Adenocarcinoma/metabolism , Cancer-Associated Fibroblasts/metabolism , Esophageal Neoplasms/metabolism , Fibroblasts/metabolism , Proteome/analysis , Adenocarcinoma/pathology , Cancer-Associated Fibroblasts/pathology , Cells, Cultured , Datasets as Topic , Esophageal Neoplasms/pathology , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Primary Cell Culture , Proteome/metabolism , Proteomics/methods , Transcriptome , Tumor Microenvironment/physiology
9.
Br J Cancer ; 119(2): 220-229, 2018 07.
Article in English | MEDLINE | ID: mdl-29991697

ABSTRACT

BACKGROUND: Immunohistochemistry (IHC) is often used in personalisation of cancer treatments. Analysis of large data sets to uncover predictive biomarkers by specialists can be enormously time-consuming. Here we investigated crowdsourcing as a means of reliably analysing immunostained cancer samples to discover biomarkers predictive of cancer survival. METHODS: We crowdsourced the analysis of bladder cancer TMA core samples through the smartphone app 'Reverse the Odds'. Scores from members of the public were pooled and compared to a gold standard set scored by appropriate specialists. We also used crowdsourced scores to assess associations with disease-specific survival. RESULTS: Data were collected over 721 days, with 4,744,339 classifications performed. The average time per classification was approximately 15 s, with approximately 20,000 h total non-gaming time contributed. The correlation between crowdsourced and expert H-scores (staining intensity × proportion) varied from 0.65 to 0.92 across the markers tested, with six of 10 correlation coefficients at least 0.80. At least two markers (MRE11 and CK20) were significantly associated with survival in patients with bladder cancer, and a further three markers showed results warranting expert follow-up. CONCLUSIONS: Crowdsourcing through a smartphone app has the potential to accurately screen IHC data and greatly increase the speed of biomarker discovery.


Subject(s)
Biomarkers, Tumor/genetics , Cell Phone , Crowdsourcing , Urinary Bladder Neoplasms/diagnosis , Female , Humans , Immunohistochemistry , Keratin-20/genetics , MRE11 Homologue Protein/genetics , Male , Middle Aged , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
10.
J Pathol ; 243(1): 37-50, 2017 09.
Article in English | MEDLINE | ID: mdl-28608476

ABSTRACT

The integrin αvß6 is up-regulated in numerous carcinomas, where expression commonly correlates with poor prognosis. αvß6 promotes tumour invasion, partly through regulation of proteases and cell migration, and is also the principal mechanism by which epithelial cells activate TGF-ß1; this latter function complicates therapeutic targeting of αvß6, since TGF-ß1 has both tumour-promoting and -suppressive effects. It is unclear how these different αvß6 functions are linked; both require actin cytoskeletal reorganization, and it is suggested that tractive forces generated during cell migration activate TGF-ß1 by exerting mechanical tension on the ECM-bound latent complex. We examined the functional relationship between cell invasion and TGF-ß1 activation in pancreatic ductal adenocarcinoma (PDAC) cells, and confirmed that both processes are αvß6-dependent. Surprisingly, we found that cellular functions could be biased towards either motility or TGF-ß1 activation depending on the presence or absence of epidermal growth factor receptor pathway substrate 8 (Eps8), a regulator of actin remodelling, endocytosis, and GTPase activation. Similar to αvß6, we found that Eps8 was up-regulated in >70% of PDACs. In complex with Abi1/Sos1, Eps8 regulated αvß6-dependent cell migration through activation of Rac1. Down-regulation of Eps8, Sos1 or Rac1 suppressed cell movement, while simultaneously increasing αvß6-dependent TGF-ß1 activation. This latter effect was modulated through increased cell tension, regulated by Rho activation. Thus, the Eps8/Abi1/Sos1 tricomplex acts as a key molecular switch altering the balance between Rac1 and Rho activation; its presence or absence in PDAC cells modulates αvß6-dependent functions, resulting in a pro-migratory (Rac1-dependent) or a pro-TGF-ß1 activation (Rho-dependent) functional phenotype, respectively. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, Neoplasm/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Cell Movement , Integrins/metabolism , Pancreatic Neoplasms/enzymology , Transforming Growth Factor beta1/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Antigens, Neoplasm/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , Humans , Integrins/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , RNA Interference , SOS1 Protein/genetics , SOS1 Protein/metabolism , Signal Transduction , Stromal Cells/enzymology , Stromal Cells/pathology , Transfection , Tumor Microenvironment , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/antagonists & inhibitors
11.
Br J Cancer ; 117(9): 1341-1348, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-28817839

ABSTRACT

BACKGROUND: We systematically assessed the prognostic and predictive value of infiltrating adaptive and innate immune cells in a large cohort of patients with advanced mesothelioma. METHODS: A tissue microarray from 302 samples was constructed. Markers of adaptive immune response in T-cells (CD8+, FOXP3+, CD4+, CD45RO+, CD3+) and B-cells (CD20+), and of innate immune response; neutrophils (NP57+), natural killer cells (CD56+) and macrophages (CD68+) were evaluated. RESULTS: We found that in the epithelioid tumours, high CD4+ and CD20+ counts, and low FOXP3+, CD68+ and NP57+ counts linked to better outcome. In the non-epithelioid group low CD8+ and low FOXP3+ counts were beneficial.On multivariate analysis low FOXP3+ remained independently associated with survival in both groups. In the epithelioid group additionally high CD4+, high CD20+, and low NP57+ counts were prognostic. CONCLUSIONS: Our data demonstrate for the first time, in predominately advanced disease, the association of key markers of adaptive and innate immunity with survival and the differential effect of histology. A better understanding of the immunological drivers of the different subtypes of mesothelioma will assist prognostication and disease-specific clinical decision-making.


Subject(s)
B-Lymphocytes/immunology , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mesothelioma/immunology , Mesothelioma/mortality , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Mesothelioma/pathology , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate
12.
Br J Cancer ; 116(2): 237-245, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27959886

ABSTRACT

BACKGROUND: Academic pathology suffers from an acute and growing lack of workforce resource. This especially impacts on translational elements of clinical trials, which can require detailed analysis of thousands of tissue samples. We tested whether crowdsourcing - enlisting help from the public - is a sufficiently accurate method to score such samples. METHODS: We developed a novel online interface to train and test lay participants on cancer detection and immunohistochemistry scoring in tissue microarrays. Lay participants initially performed cancer detection on lung cancer images stained for CD8, and we measured how extending a basic tutorial by annotated example images and feedback-based training affected cancer detection accuracy. We then applied this tutorial to additional cancer types and immunohistochemistry markers - bladder/ki67, lung/EGFR, and oesophageal/CD8 - to establish accuracy compared with experts. Using this optimised tutorial, we then tested lay participants' accuracy on immunohistochemistry scoring of lung/EGFR and bladder/p53 samples. RESULTS: We observed that for cancer detection, annotated example images and feedback-based training both improved accuracy compared with a basic tutorial only. Using this optimised tutorial, we demonstrate highly accurate (>0.90 area under curve) detection of cancer in samples stained with nuclear, cytoplasmic and membrane cell markers. We also observed high Spearman correlations between lay participants and experts for immunohistochemistry scoring (0.91 (0.78, 0.96) and 0.97 (0.91, 0.99) for lung/EGFR and bladder/p53 samples, respectively). CONCLUSIONS: These results establish crowdsourcing as a promising method to screen large data sets for biomarkers in cancer pathology research across a range of cancers and immunohistochemical stains.


Subject(s)
Biomarkers, Tumor/metabolism , Crowdsourcing/methods , Neoplasms/metabolism , Tissue Array Analysis , Translational Research, Biomedical/methods , Data Interpretation, Statistical , Humans , Image Processing, Computer-Assisted/methods , Immunohistochemistry , Patient Selection
13.
Br J Cancer ; 114(2): 134-7, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26730577

ABSTRACT

BACKGROUND: The majority (90%) of anal cancers are human papillomavirus (HPV)-driven, identified using immunochemistry for p16. Compared with HPV- patients, those with HPV+ disease generally show improved survival, although relapse rates around 25% indicate a need for further stratification of this group. METHODS: Using two cohorts of anal cancer, previously characterised for p16, we assessed the prognostic value of tumour-infiltrating lymphocytes (TILs). RESULTS: Tumour-infiltrating lymphocyte scores were used to stratify p16+ cases, where tumours with absent/low levels of TIL had a relapse-free rate of 63%, as opposed to 92% with high levels of TIL (log rank P=0.006). CONCLUSIONS: Assessment of TIL adds to p16 status in the prognosis of anal cancer following chemo-radiotherapy and provides evidence of the clinical importance of the immune response.


Subject(s)
Anus Neoplasms/therapy , Carcinoma, Squamous Cell/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Proteins/metabolism , Papillomavirus Infections/therapy , Anus Neoplasms/immunology , Anus Neoplasms/metabolism , Anus Neoplasms/virology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Chemoradiotherapy , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16 , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Papillomavirus Infections/immunology , Papillomavirus Infections/metabolism , Prognosis , Treatment Outcome
14.
Cancer Immunol Immunother ; 65(6): 651-62, 2016 06.
Article in English | MEDLINE | ID: mdl-27020682

ABSTRACT

BACKGROUND: Oesophageal adenocarcinoma (OAC) is increasingly common in the west, and survival remains poor at 10-15 % at 5 years. Immune responses are increasingly implicated as a determining factor of tumour progression. The ability of lymphocytes to recognise tumour antigens provides a mechanism for a host immune attack against cancer providing a potential treatment strategy. MATERIALS AND METHODS: Tumour infiltrating lymphocytes (TILs: CD3+, CD4+, CD8+ and FOXp3+) were assessed by immunohistochemistry using tissue microarrays in a contemporary and homogeneous cohort of OAC patients (n = 128) undergoing curative treatment. RESULTS: Multivariate analysis identified three independent prognostic factors for improved cancer-specific survival (CSS): increased CD8+ TILs (p = 0.003), completeness of resection (p < 0.0001) and lower pathological N stage (p < 0.0001). Independent prognostic factors for favourable disease-free survival included surgery-only treatment (p = 0.015), completeness of resection (p = 0.001), increased CD8+ TILs (p < 0.0001) and reduced pathological N stage (p < 0.0001). Higher levels of TILs in the pathological specimen were associated with significant pathological response to neoadjuvant chemotherapy (NAC). On multivariate analysis increased levels of CD4+ (p = 0.017) and CD8+ TILs (p = 0.005) were associated with significant local tumour regression and lymph node downstaging, respectively. DISCUSSION: Our results establish an association of TILs and survival in OAC, as seen in other solid tumours, and identify particular TIL subsets that are present at higher levels in patients who responded to NAC compared to non-responders. These findings highlight potential therapeutic strategies in EAC based on utilising the host immunological response and highlight the immune responses biomarker potential.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/mortality , Esophageal Neoplasms/immunology , Esophageal Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Biomarkers , Combined Modality Therapy , Disease Progression , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Inflammation Mediators/metabolism , Kaplan-Meier Estimate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Proportional Hazards Models , Tumor Burden
15.
J Pathol ; 235(3): 466-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25345775

ABSTRACT

Interactions between cancer cells and cancer-associated fibroblasts (CAFs) play an important role in tumour development and progression. In this study we investigated the functional role of CAFs in oesophageal adenocarcinoma (EAC). We used immunochemistry to analyse a cohort of 183 EAC patients for CAF markers related to disease mortality. We characterized CAFs and normal oesophageal fibroblasts (NOFs) using western blotting, immunofluorescence and gel contraction. Transwell assays, 3D organotypic culture and xenograft models were used to examine the effects on EAC cell function and to dissect molecular mechanisms regulating invasion. Most EACs (93%) contained CAFs with a myofibroblastic (α-SMA-positive) phenotype, which correlated significantly with poor survival [p = 0.016; HR 7. 1 (1.7-29.4)]. Primary CAFs isolated from EACs have a contractile, myofibroblastic phenotype and promote EAC cell invasion in vitro (Transwell assays, p ≤ 0.05; organotypic culture, p < 0.001) and in vivo (p ≤ 0.05). In vitro, this pro-invasive effect is modulated through the matricellular protein periostin. Periostin is secreted by CAFs and acts as a ligand for EAC cell integrins αvß3 and αvß5, promoting activation of the PI3kinase-Akt pathway. In patient samples, periostin expression at the tumour cell-stromal interface correlates with poor overall and disease-free survival. Our study highlights the importance of the tumour stroma in EAC progression. Paracrine interaction between CAF-secreted periostin and EAC-expressed integrins results in PI3 kinase-Akt activation and increased tumour cell invasion. Most EACs contain a myofibroblastic CAF-rich stroma; this may explain the aggressive, highly infiltrative nature of the disease, and suggests that stromal targeting may produce therapeutic benefit in EAC patients.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/pathology , Cell Adhesion Molecules/pharmacology , Cell Movement/drug effects , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophagus/pathology , Fibroblasts/pathology , Actins/metabolism , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/metabolism , Cells, Cultured , Cohort Studies , Disease Models, Animal , Esophageal Neoplasms/metabolism , Female , Heterografts , Humans , In Vitro Techniques , Male , Mice , Mice, SCID , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Survival Rate , Tumor Microenvironment
16.
Mod Pathol ; 28(11): 1470-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26336888

ABSTRACT

Pancreatic ductal adenocarcinoma continues to be one of the most difficult diseases to manage with one of the highest cancer mortality rates. This is due to several factors including nonspecific symptomatology and subsequent diagnosis at an advanced stage, aggressive metastatic behavior that is incompletely understood, and limited response to current therapeutic regimens. As in other cancers, there is great interest in studying the role of the tumor microenvironment in pancreatic ductal adenocarcinoma and whether components of this environment could serve as research and therapeutic targets. In particular, attention has turned toward the desmoplastic collagen-rich pancreatic ductal adenocarcinoma stroma for both biological and clinical insight. In this study, we used quantitative second harmonic generation microscopy to investigate stromal collagen organization and structure in human pancreatic ductal adenocarcinoma pathology tissues compared with non-neoplastic tissues. Collagen topology was characterized in whole-tissue microarray cores and at specific pathology-annotated epithelial-stroma interfaces representing 241 and 117 patients, respectively. We quantitatively demonstrate that a unique collagen topology exists in the periductal pancreatic ductal adenocarcinoma stroma. Specifically, collagen around malignant ducts shows increased alignment, length, and width compared with normal ducts and benign ducts in a chronic pancreatitis background. These findings indicate that second harmonic generation imaging can provide quantitative information about fibrosis that complements traditional histopathologic insights and can serve as a rich field for investigation into pathogenic and clinical implications of reorganized collagen as a pancreatic ductal adenocarcinoma disease marker.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Collagen/ultrastructure , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/pathology , Tumor Microenvironment , Area Under Curve , Humans , Immunohistochemistry , Microscopy, Confocal , ROC Curve , Sensitivity and Specificity , Tissue Array Analysis
17.
J Pathol ; 233(2): 196-208, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24573955

ABSTRACT

Aberrant Hedgehog (Hh) signalling has been reported in a number of malignancies, particularly basal cell carcinoma (BCC) of the skin. Clinical trials of Hh inhibitors are underway in many cancers, and these have produced significant clinical benefit in BCC patients, although regrowth of new, or clinically aggressive, variants, as well as development of secondary malignancies, has been reported. αvß6 integrin is expressed in many cancers, where it has been shown to correlate with an aggressive tumour phenotype and poor prognosis. We have previously reported αvß6 up-regulation in aggressive, morphoeic BCC variants, where it modulates the stromal response and induces invasion. To examine a possible link between Hh and αvß6 function, we generated BCC models, overexpressing Gli1 in immortalized keratinocytes (NTert1, HaCaT). Unexpectedly, we found that suppressing Gli1 significantly increased αvß6 expression. This promoted tumour cell motility and also stromal myofibroblast differentiation through integrin-dependent TGF-ß1 activation. Gli1 inhibited αvß6 expression by suppressing TGF-ß1-induced Smad2/3 activation, blocking a positive feedback loop maintaining high αvß6 levels. A similar mechanism was observed in AsPC1 pancreatic cancer cells expressing endogenous Gli1, suggesting a common mechanism across tumour types. In vitro findings were supported using human clinical samples, where we showed an inverse correlation between αvß6 and Gli1 expression in different BCC subtypes and pancreatic cancers. In summary, we show that expression of Gli1 and αvß6 inversely correlates in tumours in vivo, and Hh targeting up-regulates TGF-ß1/Smad2/3-dependent αvß6 expression, promoting pro-tumourigenic cell functions in vitro. These results have potential clinical significance, given the reported recurrence of BCC variants and secondary malignancies in patients treated by Hh targeting.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Basal Cell/metabolism , Cell Transformation, Neoplastic/metabolism , Hedgehog Proteins/metabolism , Integrins/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Transcription Factors/metabolism , Antigens, Neoplasm/genetics , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cell Line , Cell Movement , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Coculture Techniques , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Integrins/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors/genetics , Transfection , Transforming Growth Factor beta1/metabolism , Zinc Finger Protein GLI1
19.
Clin Cancer Res ; 30(1): 224-234, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37874322

ABSTRACT

PURPOSE: High numbers of tumor-infiltrating lymphocytes (TIL) are linked to better survival in patients with cancer. Tissue-resident memory T cells (TRM; CD8+CD103+) are recognized as a key player of anticancer immune response. To assess TRM cells in primary, metastatic, and recurrent head and neck squamous cell carcinoma (HNSCC), we developed a tissue microarray (TMA) and used multiplex IHC (MxIHC). EXPERIMENTAL DESIGN: Samples from primary tumors of 379 HNSCC cases treated at Southampton Hospitals between 2000 and 2016 were collected and analyzed. Of these, 105 cases had lymph node metastases and 82 recurrences. A TMA was generated with triplicate cores for each sample. MxIHC with a stain-and-strip approach was performed using CD8, CD103, and TIM3. Scanned slides were analyzed (digital image analysis) and quality checked (QC). RESULTS: After QC, 194 primary tumors, 76 lymph node metastases, and 65 recurrences were evaluable. Alcohol consumption was statistically significantly correlated with a reduction of TRM cells in primary tumors (nondrinker vs. heavy drinker: P = 0.0036). The known survival benefit of TRM cell infiltration in primary tumors was not found for lymph node metastasis. In recurrences, a high TRM cell number led to a favorable outcome after 12 months. The checkpoint molecule TIM3, was expressed significantly higher on TRM and non-TRM cells in the lymph node compared with primary tumors (P < 0.0001), which was also seen in recurrences (P = 0.0134 and P = 0.0007, respectively). CONCLUSIONS: We confirm the prognostic impact of TIL in primary tumors and in recurrences. TRM cell density in lymph node metastases was not linked to outcome. The role of TIM3, as a therapeutic target remains to be defined.


Subject(s)
Head and Neck Neoplasms , Neoplasms, Second Primary , Humans , Squamous Cell Carcinoma of Head and Neck , Memory T Cells , Lymphatic Metastasis , Hepatitis A Virus Cellular Receptor 2 , Immunologic Memory , Neoplasm Recurrence, Local , CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating
20.
Res Sq ; 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38496447

ABSTRACT

Two APOBEC (apolipoprotein-B mRNA editing enzyme catalytic polypeptide-like) DNA cytosine deaminase enzymes (APOBEC3A and APOBEC3B) generate somatic mutations in cancer, driving tumour development and drug resistance. Here we used single cell RNA sequencing to study APOBEC3A and APOBEC3B expression in healthy and malignant mucosal epithelia, validating key observations with immunohistochemistry, spatial transcriptomics and functional experiments. Whereas APOBEC3B is expressed in keratinocytes entering mitosis, we show that APOBEC3A expression is confined largely to terminally differentiating cells and requires Grainyhead-like transcription factor 3 (GRHL3). Thus, in normal tissue, neither deaminase appears to be expressed at high levels during DNA replication, the cell cycle stage associated with APOBEC-mediated mutagenesis. In contrast, we show that in squamous cell carcinoma tissues, there is expansion of GRHL3 expression and activity to a subset of cells undergoing DNA replication and concomitant extension of APOBEC3A expression to proliferating cells. These findings indicate a mechanism for acquisition of APOBEC3A mutagenic activity in tumours.

SELECTION OF CITATIONS
SEARCH DETAIL