1
|
Strzelec B, Chmielewski PP, Kielan W. Esophageal cancer: current status and new insights from inflammatory markers - a brief review. POLISH JOURNAL OF SURGERY 2024; 96:83-87. [PMID: 38940245 DOI: 10.5604/01.3001.0054.4523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Esophageal cancer (EC) poses a significant challenge to the healthcare system due to its profound impact on cancer-related morbidity and mortality worldwide. This malignancy ranks among the most arduous conditions confronting the surgeon. EC arises from a complex interplay of genetic predispositions and environmental factors. While the incidence of esophageal adenocarcinoma (EAC) is on the rise in the West, esophageal squamous cell carcinoma (ESCC) remains prevalent in the East. Chronic inflammation plays a pivotal role in the initiation and progression of EC. Accordingly, serum inflammatory markers, growth factors, and cytokines have been shown to be clinically useful. Thus, evaluating serum cytokine levels for EC prediction is a safe and feasible screening method. Given the aggressive nature and poor prognosis of the disease, innovative approaches to diagnosis, prognosis, and management of EC are indispensable. This review discusses the major risk factors and the current landscape of EC, with a specific focus on the potential contributions of new inflammatory markers to enhance disease management and improve patient outcomes.
Collapse
Affiliation(s)
- Bartłomiej Strzelec
- 2nd Department of General and Oncological Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Paweł Chmielewski
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Wojciech Kielan
- 2nd Department of General Surgery and Surgical Oncology, Medical University Hospital, Wroclaw, Poland
| |
Collapse
|
2
|
Peleg N, Ringel Y, Shamah S, Schmilovitz-Weiss H, Leshno M, Benjaminov F, Shinhar N, Gingold-Belfer R, Dotan I, Sapoznikov B. Development and validation of a prediction model for histologic progression in patients with nondysplastic Barrett's esophagus. Dig Endosc 2023; 35:718-725. [PMID: 36567638 DOI: 10.1111/den.14505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Patients with Barrett's esophagus (BE) are at risk of progression to esophageal adenocarcinoma (EAC). We developed a model to predict histologic progression in patients with nondysplastic BE (NDBE). METHODS A longitudinal study in three referral centers was performed between January 2010 and December 2019. As progression to low-grade dysplasia (LGD) can be considered an indication for ablative therapy, the study end-point was histopathologic progression to LGD, high-grade dysplasia, or EAC at 3 years after diagnosis. We used logistic regression to create the model. Seventy percent of the cohort were used to stem the model and the remaining 30% for internal validation. RESULTS A total of 542 patients were included, 69.4% of whom were male, mean age 62.2 years. Long-segment BE at index endoscopy was diagnosed in 20.8% of the patients. After a mean follow-up of 6.7 years, 133 patients (24.5%) had histologic progression. Our model identified a neutrophil-to-lymphocyte ratio (odds ratio [OR] 2.08, 95% confidence interval [CI] 1.77-2.32, P < 0.001), BE length (OR 1.22, 95% CI 1.09-1.36, P < 0.001), age (OR 1.03, 95% CI 1.02-1.05, P = 0.02), smoking (OR 1.66, 95% CI 1.09-2.75, P = 0.04), and renal failure (OR 1.51, 95% CI 0.93-2.43, P = 0.07) as predictors of histologic progression at 3 years. The areas under the receiver operating characteristic curves of this model were 0.88 and 0.76 in the training and validation cohorts, respectively. CONCLUSION This novel, internally validated model may predict histologic progression, even in patients with NDBE who generally have low rates of progression over time, and may contribute to enhanced patient selection for more intense surveillance programs.
Collapse
Affiliation(s)
- Noam Peleg
- Division of Gastroenterology, Rabin Medical Center, Beilinson and Hasharon Hospitals, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Ringel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Gastroenterology and Hepatology, Meir Medical Center, Kefar Sava, Israel
| | - Steven Shamah
- Division of Gastroenterology, Rabin Medical Center, Beilinson and Hasharon Hospitals, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hemda Schmilovitz-Weiss
- Division of Gastroenterology, Rabin Medical Center, Beilinson and Hasharon Hospitals, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Leshno
- Coller School of Management, Tel Aviv University, Tel Aviv, Israel
| | - Fabiana Benjaminov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Gastroenterology and Hepatology, Meir Medical Center, Kefar Sava, Israel
| | - Nadav Shinhar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Gastroenterology and Hepatology, Meir Medical Center, Kefar Sava, Israel
| | - Rachel Gingold-Belfer
- Division of Gastroenterology, Rabin Medical Center, Beilinson and Hasharon Hospitals, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Beilinson and Hasharon Hospitals, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Boris Sapoznikov
- Division of Gastroenterology, Rabin Medical Center, Beilinson and Hasharon Hospitals, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Bai J, Tan R, An Z, Xu Y. Quantitative estimation of intracellular oxidative stress in human tissues. Brief Bioinform 2022; 23:6599072. [PMID: 35653708 PMCID: PMC9294418 DOI: 10.1093/bib/bbac206] [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: 03/03/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022] Open
Abstract
Oxidative stress is known to be involved in and possibly a key driver of the development of numerous chronic diseases, including cancer. It is highly desired to have a capability to reliably estimate the level of intracellular oxidative stress as it can help to identify functional changes and disease phenotypes associated with such a stress, but the problem proves to be very challenging. We present a novel computational model for quantitatively estimating the level of oxidative stress in tissues and cells based on their transcriptomic data. The model consists of (i) three sets of marker genes found to be associated with the production of oxidizing molecules, the activated antioxidation programs and the intracellular stress attributed to oxidation, respectively; (ii) three polynomial functions defined over the expression levels of the three gene sets are developed aimed to capture the total oxidizing power, the activated antioxidation capacity and the oxidative stress level, respectively, with their detailed parameters estimated by solving an optimization problem and (iii) the optimization problem is so formulated to capture the relevant known insights such as the oxidative stress level generally goes up from normal to chronic diseases and then to cancer tissues. Systematic assessments on independent datasets indicate that the trained predictor is highly reliable and numerous insights are made based on its application results to samples in the TCGA, GTEx and GEO databases.
Collapse
Affiliation(s)
- Jun Bai
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,School of Artificial Intelligence, Jilin University, Changchun, China
| | - Renbo Tan
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,College of Computer Science and Technology, Jilin University, Changchun, China
| | - Zheng An
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, the University of Georgia, Athens, GA, USA
| | - Ying Xu
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,College of Computer Science and Technology, Jilin University, Changchun, China.,Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, the University of Georgia, Athens, GA, USA
| |
Collapse
|
4
|
Sharma T, Gupta A, Chauhan R, Bhat AA, Nisar S, Hashem S, Akhtar S, Ahmad A, Haris M, Singh M, Uddin S. Cross-talk between the microbiome and chronic inflammation in esophageal cancer: potential driver of oncogenesis. Cancer Metastasis Rev 2022; 41:281-299. [PMID: 35511379 PMCID: PMC9363391 DOI: 10.1007/s10555-022-10026-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022]
Abstract
Esophageal cancer (EC) is frequently considered a lethal malignancy and is often identified at a later stage. It is one of the major causes of cancer-related deaths globally. The conventional treatment methods like chemotherapy, radiotherapy, and surgery offer limited efficacy and poor clinical outcome with a less than 25% 5-year survival rate. The poor prognosis of EC persists despite the growth in the development of diagnostic and therapeutic modalities to treat EC. This underlines the need to elucidate the complex molecular mechanisms that drive esophageal oncogenesis. Apart from the role of the tumor microenvironment and its structural and cellular components in tumorigenesis, mounting evidence points towards the involvement of the esophageal microbiome, inflammation, and their cross-talk in promoting esophageal cancer. The current review summarizes recent research that delineates the underlying molecular mechanisms by which the microbiota and inflammation promote the pathophysiology of esophageal cancer, thus unraveling targets for potential therapeutic intervention.
Collapse
Affiliation(s)
- Tarang Sharma
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Ravi Chauhan
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India
| | - Ajaz A Bhat
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Sheema Hashem
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha, Qatar.,Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA.,Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences, New Delhi, India.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar. .,Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar. .,Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
5
|
Crosstalk between Environmental Inflammatory Stimuli and Non-Coding RNA in Cancer Occurrence and Development. Cancers (Basel) 2021; 13:cancers13174436. [PMID: 34503246 PMCID: PMC8430834 DOI: 10.3390/cancers13174436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/20/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Increasing evidence has indicated that chronic inflammatory processes have an influence on tumor occurrence and all stages of tumor development. A dramatic increase of studies into non-coding RNAs (ncRNAs) biology has shown that ncRNAs act as oncogenic drivers and tumor suppressors in various inflammation-induced cancers. Thus, this complex network of inflammation-associated cancers and ncRNAs offers targets for prevention from the malignant transformation from inflammation and treatment of malignant diseases. Abstract There is a clear relationship between inflammatory response and different stages of tumor development. Common inflammation-related carcinogens include viruses, bacteria, and environmental mutagens, such as air pollutants, toxic metals, and ultraviolet light. The expression pattern of ncRNA changes in a variety of disease conditions, including inflammation and cancer. Non-coding RNAs (ncRNAs) have a causative role in enhancing inflammatory stimulation and evading immune responses, which are particularly important in persistent pathogen infection and inflammation-to-cancer transformation. In this review, we investigated the mechanism of ncRNA expression imbalance in inflammation-related cancers. A better understanding of the function of inflammation-associated ncRNAs may help to reveal the potential of ncRNAs as a new therapeutic strategy.
Collapse
|
6
|
Elliott JA, Reynolds JV. Visceral Obesity, Metabolic Syndrome, and Esophageal Adenocarcinoma. Front Oncol 2021; 11:627270. [PMID: 33777773 PMCID: PMC7994523 DOI: 10.3389/fonc.2021.627270] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) represents an exemplar of obesity-associated carcinogenesis, with a progressive increase in EAC risk with increased body mass index. In this context, there is increased focus on visceral adipose tissue and associated metabolic dysfunction, including hypertension, diabetes mellitus and hyperlipidemia, or combinations of these in the metabolic syndrome. Visceral obesity (VO) may promote EAC via both directly impacting on gastro-esophageal reflux disease and Barrett's esophagus, as well as via reflux-independent effects, involving adipokines, growth factors, insulin resistance, and the microbiome. In this review these pathways are explored, including the impact of VO on the tumor microenvironment, and on cancer outcomes. The current evidence-based literature regarding the role of dietary, lifestyle, pharmacologic and surgical interventions to modulate the risk of EAC is explored.
Collapse
Affiliation(s)
- Jessie A Elliott
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| | - John V Reynolds
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
7
|
Abstract
Barrett’s oesophagus (BE) has been associated with an increased risk of both colorectal adenomas and colorectal cancer. A recent investigation reported a high frequency of BE in patients with adenomatous polyposis coli (APC)-associated polyposis (FAP). The aim of the present study is to evaluate the prevalence of BE in a large cohort of patients with MUTYH-associated polyposis (MAP) and APC-associated adenomatous polyposis. Patients with a genetically confirmed diagnosis of familial adenomatous polyposis (FAP) or MAP were selected and upper gastrointestinal (GI) endoscopy reports, pathology reports of upper GI biopsies were reviewed to determine the prevalence of BE in these patients. Histologically confirmed BE was found in 7 (9.7%) of 72 patients with MAP. The mean age of diagnosis was 60.2 years (range 54.1–72.4 years). Two patients initially diagnosed with low grade dysplasia showed fast progression into high grade dysplasia and esophageal cancer, respectively. Only 4 (1.4%) of 365 patients with FAP were found to have pathologically confirmed BE. The prevalence of BE in patients with MAP is much higher than reported in the general population. We recommend that upper GI surveillance of patients with MAP should not only focus on the detection of gastric and duodenal adenomas but also on the presence of BE.
Collapse
|
8
|
Zhao M, Wang J, Yuan M, Ma Z, Bao Y, Hui Z. Multivariate gene expression-based survival predictor model in esophageal adenocarcinoma. Thorac Cancer 2020; 11:2896-2908. [PMID: 32869505 PMCID: PMC7529573 DOI: 10.1111/1759-7714.13626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
Background Despite the recent development of molecular‐targeted treatment and immunotherapy, survival of patients with esophageal adenocarcinoma (EAC) with poor prognosis is still poor due to lack of an effective biomarker. In this study, we aimed to explore the ceRNA and construct a multivariate gene expression predictor model using data from The Cancer Genome Atlas (TCGA) to predict the prognosis of EAC patients. Methods We conducted differential expression analysis using mRNA, miRNA and lncRNA transciptome data from EAC and normal patients as well as corresponding clinical information from TCGA database, and gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of those unique differentially expressed mRNAs using the Integrate Discovery Database (DAVID) database. We then constructed the lncRNA‐miRNA‐mRNA competing endogenous RNA (ceRNA) network of EAC and used Cox proportional hazard analysis to generate a multivariate gene expression predictor model. We finally performed survival analysis to determine the effect of differentially expressed mRNA on patients' overall survival and discover the hub gene. Results We identified a total of 488 lncRNAs, 33 miRNAs, and 1207 mRNAs with differentially expressed profiles. Cox proportional hazard analysis and survival analysis using the ceRNA network revealed four genes (IL‐11, PDGFD, NPTX1, ITPR1) as potential biomarkers of EAC prognosis in our predictor model, and IL‐11 was identified as an independent prognostic factor. Conclusions In conclusion, we identified differences in the ceRNA regulatory networks and constructed a four–gene expression‐based survival predictor model, which could be referential for future clinical research.
Collapse
Affiliation(s)
- Maoyuan Zhao
- Department of Radiation Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jingsong Wang
- State Key Laboratory of Molecular Oncology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meng Yuan
- Department of Radiation Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zeliang Ma
- Department of Radiation Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongxin Bao
- Department of Radiation Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhouguang Hui
- Department of Radiation Oncology, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of VIP Medical Services, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Xie SH, Rabbani S, Ness-Jensen E, Lagergren J. Circulating Levels of Inflammatory and Metabolic Biomarkers and Risk of Esophageal Adenocarcinoma and Barrett Esophagus: Systematic Review and Meta-analysis. Cancer Epidemiol Biomarkers Prev 2020; 29:2109-2118. [PMID: 32855267 DOI: 10.1158/1055-9965.epi-20-0572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/05/2020] [Accepted: 08/21/2020] [Indexed: 11/16/2022] Open
Abstract
Associations between circulating levels of obesity-related biomarkers and risk of esophageal adenocarcinoma and Barrett esophagus have been reported, but the results are inconsistent. A literature search until October 2018 in MEDLINE and EMBASE was performed. Pooled ORs with 95% confidence intervals (CI) were estimated for associations between 13 obesity-related inflammatory and metabolic biomarkers and risk of esophageal adenocarcinoma or Barrett esophagus using random effect meta-analyses. Among 7,641 studies, 19 were eligible for inclusion (12 cross-sectional, two nested case-control, and five cohort studies). Comparing the highest versus lowest categories of circulating biomarker levels, the pooled ORs were increased for leptin (OR, 1.68; 95% CI, 0.95-2.97 for Barrett esophagus), glucose (OR, 1.12; 95% CI, 1.03-1.22 for esophageal adenocarcinoma), insulin (OR, 1.47; 95% CI, 1.06-2.00 for Barrett esophagus), C-reactive protein (CRP; OR, 2.06; 95% CI, 1.28-3.31 for esophageal adenocarcinoma), IL6 (OR, 1.50; 95% CI, 1.03-2.19 for esophageal adenocarcinoma), and soluble TNF receptor 2 (sTNFR-2; OR, 3.16; 95% CI, 1.76-5.65 for esophageal adenocarcinoma). No associations were identified for adiponectin, ghrelin, insulin-like growth factor 1, insulin-like growth factor-binding protein 3, triglycerides, IL8, or TNFα. Higher circulating levels of leptin, glucose, insulin, CRP, IL6, and sTNFR-2 may be associated with an increased risk of esophageal adenocarcinoma or Barrett esophagus. More prospective studies are required to identify biomarkers that can help select high-risk individuals for targeted prevention and early detection.
Collapse
Affiliation(s)
- Shao-Hua Xie
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Sirus Rabbani
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eivind Ness-Jensen
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Medical Department, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Jesper Lagergren
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
10
|
Karstens KF, Kempski J, Giannou AD, Freiwald E, Reeh M, Tachezy M, Izbicki JR, Lohse AW, Gagliani N, Huber S, Pelczar P. Systemic interleukin 10 levels indicate advanced stages while interleukin 17A levels correlate with reduced survival in esophageal adenocarcinomas. PLoS One 2020; 15:e0231833. [PMID: 32298379 PMCID: PMC7162521 DOI: 10.1371/journal.pone.0231833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 03/29/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Reflux promotes esophageal adenocarcinomas (EAC) creating a chronic inflammatory environment. EAC show an increasing incidence in the Western World and median survival rates are still low. The main reasons for poor prognosis despite new multimodal therapies are diagnosis of EACs at an already advanced stage and distant metastases. Hence, we wanted to investigate the presence of systemic inflammatory interleukins (IL) and their impact on patient prognosis. Material and methods Systemic expression levels of pro- and anti-inflammatory markers (IL-2, IL-4, IL-6, IL-10, IL-17A and IL-22) in the sera of 43 EAC patients without neoadjuvant radiochemotherapy were measured by flow cytometric analysis. A correlation to clinicopathological data was performed. Log-rank and Cox regression analysis were used to investigate the impact on patient survival. 43 sera of age and gender matched healthy volunteers were used as controls. Results Increased systemic IL-6 (p = 0.044) and lower IL-17A (p = 0.002) levels were found in EAC patients as opposed to controls. A correlation of IL-10 levels with an increased T stage was found (p = 0.020). Also, systemic IL-10 levels were highly elevated in patients with distant metastasis (p<0.001). However, only systemic IL-17A levels had an influence on patient survival in multivariate analysis. Conclusion Systemic IL-6 levels are increased, while IL-17A levels are reduced in EAC patients compared to healthy controls. In addition, circulating IL-10 might help to identify patients with advanced disease and high IL-17A might indicate a limited prognosis.
Collapse
Affiliation(s)
- Karl-Frederick Karstens
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Kempski
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasios D. Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Erik Freiwald
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W. Lohse
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| | - Penelope Pelczar
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Zhu J, Ling Y, Mi S, Chen H, Fan J, Cai S, Fan C, Shen Q, Li Y. Association between dietary inflammatory index and upper aerodigestive tract cancer risk: A systematic review and dose-response meta-analysis. Oral Oncol 2020; 103:104587. [PMID: 32050153 DOI: 10.1016/j.oraloncology.2020.104587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The relationship between dietary inflammatory index (DII) and upper aerodigestive tract (UADT) cancer risk have been investigated in a growing number of epidemiological studies. However, their findings were inconsistent, and no systematic review or meta-analysis has been conducted up to now. This meta-analysis was carried out to examine potential dose-response relationship between DII score and UADT cancer risk. MATERIAL AND METHODS A systematic search was conducted for relevant studies in PubMed and Web of Science up to March 28, 2019. Categorical meta-analysis as well as linear and non-linear dose-response meta-analysis were performed to evaluate association between DII and UADT cancer risk. RESULTS Nine case-control studies with a total of 4138 cases and 15,326 healthy controls were eligible in the present meta-analysis. The pooled odds ratios (ORs) of UADT cancer risk were 2.07 [95% confidence interval (CI): 1.82, 2.35] for the highest DII score compared with the lowest and 1.53 (95% CI: 1.39, 1.69) for higher DII score compared with lower score, respectively. Furthermore, a one-unit increment in DII score was associated with an increased risk of 18% for UADT cancers (OR: 1.18; 95% CI: 1.15, 1.21). An upward trend towards a positive association between elevated DII score and UADT cancer risk was also observed in non-linear dose-response meta-analysis. CONCLUSIONS The present meta-analysis provides evidence of highly pro-inflammatory diets that might increase risk of UADT cancers. Therefore, reducing pro-inflammatory components in diets should be considered to prevent and control UADT cancers.
Collapse
Affiliation(s)
- Jiahao Zhu
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuxiao Ling
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shuai Mi
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hanzhu Chen
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiayao Fan
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shaofang Cai
- Department of Science and Education, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Chunhong Fan
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qing Shen
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yingjun Li
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Neutrophil-Lymphocyte Ratio as a Marker of Progression from Non-Dysplastic Barrett's Esophagus to Esophageal Adenocarcinoma: a Cross-Sectional Retrospective Study. J Gastrointest Surg 2020; 24:8-18. [PMID: 31745889 DOI: 10.1007/s11605-019-04456-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Immune imbalance and inflammation have been suggested as key factors of Barrett's esophagus (BE) pathway towards adenocarcinoma. The neutrophil-lymphocyte ratio (NLR) indirectly reflects the relation between innate and adaptive immune systems and has been studied in premalignant conditions as a biomarker for cancer diagnosis. Our aim was to investigate if increasing values of NLR correlated with advancing stages of BE progression to dysplasia and neoplasia. METHODS We retrospectively analyzed data of patients with biopsies reporting BE between 2013 and 2017 and with a complete blood count within 6 months from the endoscopy, as well as patients with esophageal adenocarcinoma (EAC). NLR was calculated as neutrophil count/lymphocyte count. Cases (n = 113) were classified as non-dysplastic BE (NDBE, n = 72), dysplastic BE (DBE, n = 11) and EAC (n = 30). RESULTS NLR progressively increased across groups (NDBE, 1.92 ± 0.7; DBE, 2.92 ± 1.1; EAC 4.54 ± 2.9), with a significant correlation between its increasing value and the presence of dysplasia or neoplasia (r = 0.53, p < 0.001). NLR > 2.27 was able to diagnose EAC with 80% sensitivity and 71% specificity (area under the curve = 0.8). CONCLUSION NLR correlates with advancing stages of BE progression, a finding that reinforces the role of immune imbalance in EAC carcinogenesis and suggests a possible use of this marker for risk stratification on surveillance strategies.
Collapse
|
13
|
Münch NS, Fang HY, Ingermann J, Maurer HC, Anand A, Kellner V, Sahm V, Wiethaler M, Baumeister T, Wein F, Einwächter H, Bolze F, Klingenspor M, Haller D, Kavanagh M, Lysaght J, Friedman R, Dannenberg AJ, Pollak M, Holt PR, Muthupalani S, Fox JG, Whary MT, Lee Y, Ren TY, Elliot R, Fitzgerald R, Steiger K, Schmid RM, Wang TC, Quante M. High-Fat Diet Accelerates Carcinogenesis in a Mouse Model of Barrett's Esophagus via Interleukin 8 and Alterations to the Gut Microbiome. Gastroenterology 2019; 157:492-506.e2. [PMID: 30998992 PMCID: PMC6662596 DOI: 10.1053/j.gastro.2019.04.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 04/03/2019] [Accepted: 04/06/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS Barrett's esophagus (BE) is a precursor to esophageal adenocarcinoma (EAC). Progression from BE to cancer is associated with obesity, possibly due to increased abdominal pressure and gastroesophageal reflux disease, although this pathogenic mechanism has not been proven. We investigated whether environmental or dietary factors associated with obesity contribute to the progression of BE to EAC in mice. METHODS Tg(ED-L2-IL1RN/IL1B)#Tcw mice (a model of BE, called L2-IL1B mice) were fed a chow (control) or high-fat diet (HFD) or were crossbred with mice that express human interleukin (IL) 8 (L2-IL1B/IL8 mice). Esophageal tissues were collected and analyzed for gene expression profiles and by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry. Organoids were established from BE tissue of mice and cultured with serum from lean or obese individuals or with neutrophils from L2-IL1B mice. Feces from mice were analyzed by 16s ribosomal RNA sequencing and compared to 16s sequencing data from patients with dysplasia or BE. L2-IL1B were mice raised in germ-free conditions. RESULTS L2-IL1B mice fed an HFD developed esophageal dysplasia and tumors more rapidly than mice fed the control diet; the speed of tumor development was independent of body weight. The acceleration of dysplasia by the HFD in the L2-IL1B mice was associated with a shift in the gut microbiota and an increased ratio of neutrophils to natural killer cells in esophageal tissues compared with mice fed a control diet. We observed similar differences in the microbiomes from patients with BE that progressed to EAC vs patients with BE that did not develop into cancer. Tissues from dysplasias of L2-IL1B mice fed the HFD contained increased levels of cytokines that are produced in response to CXCL1 (the functional mouse homolog of IL8, also called KC). Serum from obese patients caused organoids from L2-IL1B/IL8 mice to produce IL8. BE tissues from L2-IL1B mice fed the HFD and from L2-IL1B/IL8 mice contained increased numbers of myeloid cells and cells expressing Cxcr2 and Lgr5 messenger RNAs (epithelial progenitors) compared with mice fed control diets. BE tissues from L2-IL1B mice raised in germ-free housing had fewer progenitor cells and developed less dysplasia than in L2-IL1 mice raised under standard conditions; exposure of fecal microbiota from L2-IL1B mice fed the HFD to L2-IL1B mice fed the control diet accelerated tumor development. CONCLUSIONS In a mouse model of BE, we found that an HFD promoted dysplasia by altering the esophageal microenvironment and gut microbiome, thereby inducing inflammation and stem cell expansion, independent of obesity.
Collapse
Affiliation(s)
- Natasha Stephens Münch
- Department of Internal Medicine, Technical University of Munich, Germany,Chair of Molecular Nutritional Medicine, Technical University of Munich, Germany
| | - Hsin-Yu Fang
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Jonas Ingermann
- Department of Internal Medicine, Technical University of Munich, Germany,Chair of Molecular Nutritional Medicine, Technical University of Munich, Germany
| | - H. Carlo Maurer
- Department of Internal Medicine, Technical University of Munich, Germany,Irvine Cancer Research Center, Columbia University, New York, USA
| | - Akanksha Anand
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Victoria Kellner
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Vincenz Sahm
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Maria Wiethaler
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Theresa Baumeister
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Frederik Wein
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Henrik Einwächter
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Florian Bolze
- Chair of Molecular Nutritional Medicine, Technical University of Munich, Germany,EKFZ – Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Germany,ZIEL – Institute of Food & Health, Technical University of Munich, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University of Munich, Germany,EKFZ – Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Germany,ZIEL – Institute of Food & Health, Technical University of Munich, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology; Technical University of Munich, Germany
| | - Maria Kavanagh
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Richard Friedman
- Irvine Cancer Research Center, Columbia University, New York, USA
| | | | | | | | | | - James G. Fox
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mark T. Whary
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoomi Lee
- Irvine Cancer Research Center, Columbia University, New York, USA
| | - Tony Y. Ren
- Irvine Cancer Research Center, Columbia University, New York, USA
| | | | | | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Germany
| | - Roland M. Schmid
- Department of Internal Medicine, Technical University of Munich, Germany
| | - Timothy C. Wang
- Irvine Cancer Research Center, Columbia University, New York, USA
| | - Michael Quante
- Department of Internal Medicine, Technical University of Munich, Germany.
| |
Collapse
|
14
|
Cook MB, Barnett MJ, Bock CH, Cross AJ, Goodman PJ, Goodman GE, Haiman CA, Khaw KT, McCullough ML, Newton CC, Boutron-Ruault MC, Lund E, Rutegård M, Thornquist MD, Spriggs M, Giffen C, Freedman ND, Kemp T, Kroenke CH, Le Marchand L, Park JY, Simon M, Wilkens LR, Pinto L, Hildesheim A, Campbell PT. Prediagnostic circulating markers of inflammation and risk of oesophageal adenocarcinoma: a study within the National Cancer Institute Cohort Consortium. Gut 2019; 68:960-968. [PMID: 30121626 PMCID: PMC6379150 DOI: 10.1136/gutjnl-2018-316678] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/02/2018] [Accepted: 08/02/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Cross-sectional data indicate that systemic inflammation is important in oesophageal adenocarcinoma. We conducted a prospective study to assess whether prediagnostic circulating markers of inflammation were associated with oesophageal adenocarcinoma and to what extent they mediated associations of obesity and cigarette smoking with cancer risk. DESIGN This nested case-control study included 296 oesophageal adenocarcinoma cases and 296 incidence density matched controls from seven prospective cohort studies. We quantitated 69 circulating inflammation markers using Luminex-based multiplex assays. Conditional logistic regression models estimated associations between inflammation markers and oesophageal adenocarcinoma, as well as direct and indirect effects of obesity and smoking on risk of malignancy. RESULTS Soluble tumour necrosis factor receptor 2 (sTNFR2) (ORsquartile 4 vs 1=2.67, 95% CI 1.52 to 4.68) was significantly associated with oesophageal adenocarcinoma. Additional markers close to the adjusted significance threshold included C reactive protein, serum amyloid A, lipocalin-2, resistin, interleukin (IL) 3, IL17A, soluble IL-6 receptor and soluble vascular endothelial growth factor receptor 3. Adjustment for body mass index, waist circumference or smoking status slightly attenuated biomarker-cancer associations. Mediation analysis indicated that sTNFR2 may account for 33% (p=0.005) of the effect of waist circumference on oesophageal adenocarcinoma risk. Resistin, plasminogen activator inhibitor 1, C reactive protein and serum amyloid A were also identified as potential mediators of obesity-oesophageal adenocarcinoma associations. For smoking status, only plasminogen activator inhibitor 1 was a nominally statistically significant (p<0.05) mediator of cancer risk. CONCLUSION This prospective study provides evidence of a link between systemic inflammation and oesophageal adenocarcinoma risk. In addition, this study provides the first evidence that indirect effects of excess adiposity and cigarette smoking, via systemic inflammation, increase the risk of oesophageal adenocarcinoma.
Collapse
Affiliation(s)
- Michael B. Cook
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, Maryland, USA
| | - Matthew J. Barnett
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Cathryn H. Bock
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Amanda J. Cross
- Department of Epidemiology and Biostatistics, Imperial College London, UK
| | - Phyllis J. Goodman
- Southwest Oncology Group (SWOG) Statistics & Data Management Center (SDMC), Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Gary E. Goodman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- Swedish Medical Center, Swedish Cancer Institute, Seattle, WA 98104
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, United Kingdom
| | - Marjorie L. McCullough
- Behavioral and Epidemiology Research Program, American Cancer Society Inc., Atlanta, GA 30303, USA
| | - Christine C. Newton
- Behavioral and Epidemiology Research Program, American Cancer Society Inc., Atlanta, GA 30303, USA
| | - Marie-Christine Boutron-Ruault
- CESP, Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, 94805, Villejuif, France
- Generations and Health, Gustave Roussy, F-94805, Villejuif, France
| | - Eiliv Lund
- Department of Community Medicine, UiT The Arctic University of Norway, 9037 Tromsø, Norway
| | - Martin Rutegård
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Mark D. Thornquist
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Michael Spriggs
- Information Management Services (IMS), Rockville, MD 20852, USA
| | - Carol Giffen
- Information Management Services (IMS), Rockville, MD 20852, USA
| | - Neal D. Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, Maryland, USA
| | - Troy Kemp
- Human Papilloma Virus (HPV) Immunology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Candyce H. Kroenke
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo St., Honolulu, HI 96817, USA
| | - Jin Young Park
- Prevention and Implementation Group, International Agency for Research on Cancer, Lyon, France
| | - Michael Simon
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Lynne R. Wilkens
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - Ligia Pinto
- Information Management Services (IMS), Rockville, MD 20852, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, Maryland, USA
| | - Peter T. Campbell
- Behavioral and Epidemiology Research Program, American Cancer Society Inc., Atlanta, GA 30303, USA
| |
Collapse
|
15
|
Wang XB, Wu DJ, Chen WP, Liu J, Ju YJ. Impact of radiotherapy on immunological parameters, levels of inflammatory factors, and clinical prognosis in patients with esophageal cancer. JOURNAL OF RADIATION RESEARCH 2019; 60:353-363. [PMID: 31034571 PMCID: PMC6530619 DOI: 10.1093/jrr/rrz006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/05/2018] [Indexed: 05/10/2023]
Abstract
The aim of this study was to observe dynamic changes in immunological parameters and levels of inflammatory factors from pre-radiotherapy to post-radiotherapy in patients with esophageal cancer, and to evaluate the related clinical prognosis. In all, 110 patients with esophageal cancer who underwent radiotherapy were enrolled. Before radiotherapy, post-radiotherapy, and 3 months after radiotherapy, the percentages of T lymphocyte subsets and natural killer (NK) cells in peripheral blood were detected using flow cytometry. The levels of serum inflammatory factors were measured with the enzyme-linked immunosorbent assay (ELISA). Thirty peripheral blood samples from healthy people were similarly analysed as the control. Before radiotherapy, the percentages of CD4+ and CD8+ T cells and NK cells, and the CD4+/CD8+ rate in esophageal cancer patients were significantly different from those in the healthy control group (P < 0.001); the levels of inflammatory factors were increased significantly (P < 0.001). The percentages of the above cells and the levels of inflammatory factors also differed statistically significantly between pre- and post-radiotherapy (P < 0.001) in the esophageal cancer patients. Three months after radiotherapy, the percentages of CD3+ (P = 0.453), CD4+ (P = 0.108), and CD8+ T cells (P = 0.163) and NK cells (P = 0.103) had recovered to the level before radiotherapy; and the levels of TNF-α (P = 0.101), IL-6 (P = 0.302) and IL-8 (P = 0.250) were also restored. After radiotherapy, alterations in immunological parameters were associated with the irradiation volume and the myelosuppression condition. Patients with recovered immunological parameters showed a longer median survival time than those with poor recovery of immunological parameters. For esophageal cancer patients who were immunosuppressive and had an activated inflammatory response before radiotherapy, radiotherapy aggravated these symptoms, and this aggravation was positively associated with myelosuppression and irradiation volume. In addition, recovery of the immunological parameters indicated better prognosis.
Collapse
Affiliation(s)
- Xiao-Bo Wang
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Di-Jun Wu
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Wei-Ping Chen
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Jian Liu
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Yong-Jian Ju
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
- Corresponding author: Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong 226001, P.R. China. Tel/Fax: +86-0513-85061155.
| |
Collapse
|
16
|
Kresty LA, Fromkes JJ, Frankel WL, Hammond CD, Seeram NP, Baird M, Stoner GD. A phase I pilot study evaluating the beneficial effects of black raspberries in patients with Barrett's esophagus. Oncotarget 2018; 9:35356-35372. [PMID: 30450163 PMCID: PMC6219678 DOI: 10.18632/oncotarget.10457] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/23/2016] [Indexed: 12/11/2022] Open
Abstract
Black raspberries inhibit a broad range of cancers in preclinical models which has led to clinical evaluations targeting premalignant lesions of the colon, oral cavity and esophagus. A phase I pilot study was conducted in twenty Barrett's esophagus (BE) patients to investigate the effect of lyophilized black raspberries (LBR) on urinary metabolites and markers of lipid peroxidation, DNA damage and tissue markers of cellular proliferation, detoxification, and inflammation. Surveys, biopsies, blood and urine samples were collected before and after 6 months of LBR treatment (32 or 45 g). LBR significantly reduced urinary excretion of 8-epi-prostaglandin F2α, a marker of lipid peroxidation linked to oxidative stress and free radical damage. Urinary levels of the ellagitannin metabolites, urolithin A-glucuronide, urolithin A-sulfate and dimethylellagic acid glucuronide were significantly increased following 12 and 26 weeks of LBR consumption and may prove useful as indicators of compliance in future clinical studies. Immunohistochemical staining of BE biopsies following LBR treatment showed significant increases in mean GST-pi levels, with 55.6% of subjects responding favorably. In summary, LBR significantly decreased urinary lipid peroxidation levels and significantly increased GST-pi, a marker of detoxification, in BE epithelium. Still, LBR may need to be formulated differently, administered at higher concentrations or multiple times a day to increase efficacy.
Collapse
Affiliation(s)
- Laura A Kresty
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John J Fromkes
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Cynthia D Hammond
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Navindra P Seeram
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Maureen Baird
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Gary D Stoner
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
17
|
Leukaemia inhibitory factor is associated with treatment resistance in oesophageal adenocarcinoma. Oncotarget 2018; 9:33634-33647. [PMID: 30263091 PMCID: PMC6154751 DOI: 10.18632/oncotarget.25950] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023] Open
Abstract
Oesophageal cancer is an aggressive disease with a poor 5 year survival rate of <20% of diagnosed patients. Unfortunately, only 20-30% Oesophageal Adenocarinoma (OAC) patients show a beneficial response to neoadjuvant therapy (neoCT). Inflammation influences OAC given the increased risk of cancer development and poor outcome for obese patients where altered secretion of adipokines and cytokines from adipose tissue contributes a pro-tumourigenic environment. We carried out a large proteomics screen of 184 proteins to compare the inflammatory and oncogenic profiles of an isogenic radioresistant in-vitro model of OAC. We found that leukaemia inhibitory factor (LIF), an IL-6 type cytokine, was significantly elevated in radioresistant OAC cells (p=0.007). Furthermore, significantly higher circulating levels of LIF were present in the serum from treatment-naive OAC patients who had a subsequent poor pathological response to neo-adjuvant therapy, (p=0.037). Quantitative PCR analysis revealed expression of LIF receptor (LIFR) may function as a predictive indicator of response to neo-adjuvant chemoradiation therapy in OAC. LIF was demonstrated to be actively secreted from human OAC treatment-naïve biopsies and significantly correlated with the secretion of bFGF, VEGF-A and IL-8 (p<0.05, R=1), (p<0.05, R=0.9429), and (p<0.05, R=1) respectively. Importantly, LIF secretion negatively correlated with tumour infiltrating lymphocytes in pre-treatment OAC patient biopsies, (r=-0.8783, p=0.033). Elevated circulating LIF is a marker of poor response to neo-adjuvant treatment in OAC and secretion of this chemokine from the tumour is tightly linked with pro-tumourigenic mediators including bFGF, VEGF-A and IL-8. Targeting this pathway may be a novel mechanism enhance neoadjuvant treatment responses in OAC.
Collapse
|
18
|
Gào X, Brenner H, Holleczek B, Cuk K, Zhang Y, Anusruti A, Xuan Y, Xu Y, Schöttker B. Urinary 8-isoprostane levels and occurrence of lung, colorectal, prostate, breast and overall cancer: Results from a large, population-based cohort study with 14 years of follow-up. Free Radic Biol Med 2018; 123:20-26. [PMID: 29778463 DOI: 10.1016/j.freeradbiomed.2018.05.065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Urinary 8-isoprostane is an established biomarker for lipid peroxidation. However, the association between its pre-diagnostic levels and cancer incidence has rarely been evaluated. METHODS 8793 older adults from the German ESTHER cohort were followed up for cancer incidence by cancer registry data. A directed acyclic graph was utilized to identify potential confounders. Multivariate Cox regression models were applied to estimate hazard ratios (HRs) and 95% confidence intervals (95% CI). RESULTS During 14-year follow-up, 1540 incident cancer cases, including 207 lung, 196 colorectal, 218 breast and 245 prostate cancer cases were detected. 8-isoprostane concentrations were positively associated with lung cancer, but not with cancer at the other sites. The HR (95% CI) for the association with lung cancer was 1.61 (1.10, 2.34) for comparison of the top with bottom tertile in total population. The association of 8-isoprostane levels with lung cancer persisted after the adjustment for smoking and other potential confounders and was multiplicative to the effect of smoking. However, 8-isoprostane levels did not improve lung cancer prediction when added to a model containing age, sex and smoking. A protective association of increasing 8-isoprostane levels was observed for prostate cancer incidence but this association was only statistically significant among current smokers. DISCUSSION Our findings suggest that lipid peroxidation is involved in the development of lung cancer. However, high oxidative stress may be a protective factor for prostate cancer, especially among current smokers.
Collapse
Affiliation(s)
- Xīn Gào
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, Heidelberg University, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Katarina Cuk
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany
| | - Yan Zhang
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany
| | - Ankita Anusruti
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, Heidelberg University, Germany
| | - Yang Xuan
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, Heidelberg University, Germany
| | - Yiwei Xu
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ben Schöttker
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, Heidelberg University, Germany; Institute of Health Care and Social Sciences, FOM University, Essen, Germany.
| |
Collapse
|
19
|
Abe M, Shivappa N, Ito H, Oze I, Abe T, Shimizu Y, Hasegawa Y, Kiyohara C, Nomura M, Ogawa Y, Hebert JR, Matsuo K. Dietary inflammatory index and risk of upper aerodigestive tract cancer in Japanese adults. Oncotarget 2018; 9:24028-24040. [PMID: 29844870 PMCID: PMC5963633 DOI: 10.18632/oncotarget.25288] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 04/13/2018] [Indexed: 01/10/2023] Open
Abstract
Background The inflammatory potential of diet that has been shown to be associated with cancer risk. We examined the association between dietary inflammatory potential as measured by the dietary inflammatory index (DII®) and risk of upper aerodigestive tract cancers in a Japanese case-control study. Results A positive association was observed between increasing DII scores and overall upper aerodigestive tract cancers, and across anatomic subsites. For upper aerodigestive tract cancers, the ORQ4vsQ1 = 1.73 (95% CI: 1.37–2.20); head and neck cancer, the ORQ4vsQ1 was 1.92 (95% CI: 1.42–2.59); and for esophageal cancer, the ORQ4vsQ1 was1.71 (95% CI: 1.54–1.90). Risks for hypopharyngeal and nasopharyngeal cancers were greatly elevated: (ORQ4vsQ1 = 4.05 (95% CI: 1.24–13.25) for hypopharyngeal cancer and ORQ4vsQ1 = 4.99 (95% CI: 1.14–21.79) for nasopharyngeal cancer. Conclusion A more pro-inflammatory diet was associated with an elevated risk of upper aerodigestive tract cancers after accounting for important confounders. All anatomic subsites, except larynx, showed the consistently elevated risk with increasing DII score. Those subsites with known etiological associations with persistent infection showed the largest elevation in risk. These results warrant further evaluation in future studies. Materials and Methods This is a case-control study of 1,028 cases and 3,081 age- and sex-matched non-cancer controls recruited at Aichi Cancer Center. DII scores were computed based on estimates of macro- and micro-nutrients from a self-administered food frequency questionnaire. Scores were further categorized into quartiles (based on the distribution in controls). Conditional logistic regression models were fit to estimate odds ratio (OR) and 95% confidence intervals (CIs) adjusted for smoking, ethanol consumption, alcohol flushing, number of teeth, and occupation group.
Collapse
Affiliation(s)
- Makiko Abe
- Department of Preventive Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, 812-8582, Japan.,Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan.,Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Nitin Shivappa
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC, 29208, USA.,Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA.,Connecting Health Innovations LLC, Columbia, SC, 29201, USA
| | - Hidemi Ito
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan.,Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Isao Oze
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Tetsuya Abe
- Department of Gastrointestinal Surgery, Aichi Cancer Center Hospital, Nagoya, 464-8681, Japan
| | - Yasuhiro Shimizu
- Department of Gastrointestinal Surgery, Aichi Cancer Center Hospital, Nagoya, 464-8681, Japan
| | - Yasuhisa Hasegawa
- Department of Head and Neck Surgery, Aichi Cancer Center Hospital, Nagoya, 464-8681, Japan
| | - Chikako Kiyohara
- Department of Preventive Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Masatoshi Nomura
- Department of Endocrinology and Metabolism, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan.,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - James R Hebert
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC, 29208, USA.,Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA.,Connecting Health Innovations LLC, Columbia, SC, 29201, USA
| | - Keitaro Matsuo
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan.,Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
20
|
Caughron B, Yang Y, Young MRI. Role of IL-23 signaling in the progression of premalignant oral lesions to cancer. PLoS One 2018; 13:e0196034. [PMID: 29664967 PMCID: PMC5903614 DOI: 10.1371/journal.pone.0196034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/05/2018] [Indexed: 12/26/2022] Open
Abstract
Mice bearing carcinogen-induced premalignant oral lesions were previously shown to have a pro-inflammatory phenotype, which is replaced with an immune inhibitory phenotype as lesions progress to cancer. Since Th17 cells are prominent at the premalignant lesion state and their levels are supported by IL-23, studies used mice that were IL-23 receptor deficient (IL-23R KO) to determine the requirement for IL-23 signaling in the immunological and clinical status of mice with premalignant oral lesions. The results showed a dependence on IL-23 signaling for the pro-inflammatory state of mice with oral lesions as levels of IL-2, IFN-γ, IL-6, IL-17 and TNF-α were elevated in wildtype mice with premalignant oral lesions, but not in IL-23R KO mice. In contrast, as lesions progressed to cancer, the pro-inflammatory phenotype subsided and was replaced with the inhibitory mediator IL-10 and with Treg cells in wildtype mice, although not in IL-23R KO mice. Clinically, early progression of premalignant oral lesions to cancer was enhanced in IL-23R KO mice compared to progression in wildtype mice. These results show the importance of IL-23 signaling in both the pro-inflammatory phenotype characteristic of premalignant oral lesions and the inhibitory phenotype as lesions progress to cancer.
Collapse
Affiliation(s)
- Blaine Caughron
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC, United States of America
| | - Yi Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States of America
| | - M. Rita I. Young
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC, United States of America
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, United States of America
- * E-mail:
| |
Collapse
|
21
|
Tiasto V, Mikhailova V, Gulaia V, Vikhareva V, Zorin B, Kalitnik A, Kagansky A. Esophageal cancer research today and tomorrow: Lessons from algae and other perspectives. AIMS GENETICS 2018; 5:75-90. [PMID: 31435514 PMCID: PMC6690251 DOI: 10.3934/genet.2018.1.75] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/01/2018] [Indexed: 12/16/2022]
Abstract
Esophageal cancer is an increasing concern due to poor prognosis, aggressive disease modalities, and a lack of efficient therapeutics. The two types of esophageal cancer: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) are responsible for an estimated 450,000 annual deaths, with over 457,000 new patients diagnosed in 2015, making it the eighth most prevalent and the 10th most fatal cancer worldwide. As esophageal cancer prevalence continues to increase, and so does the pressing need for the development of new and effective strategies for the early diagnostics, prevention, and treatment of this cancer, as well for building the innovative research tools to understand the affected molecular mechanisms. This short review summarizes the current statistics and recent research of the problems and solutions related to the esophageal cancer, and offer a brief overview of its epidemiology, molecular alterations, and existing biomedical tools. We will discuss currently available research tools and discuss selected approaches we deem relevant to find new model systems and therapies for the future with the special focus on novel opportunities presented by the unique molecules found in algae, namely carbohydrates and lipids. Their remarkable chemical variability is connected to their striking structural and functional properties, which combined with the relative novelty of these compounds to cancer biology, warrants interest of the wide biomedical community to these molecules, especially in the esophageal cancer theory and practice.
Collapse
Affiliation(s)
- Vladlena Tiasto
- Centre for Genomic and Regenerative Medicine, School of Biomedicine, FEFU, 8 Sukhanova str, Vladivostok, Primorsky region, 690950, Russian Federation
| | - Valeriia Mikhailova
- Centre for Genomic and Regenerative Medicine, School of Biomedicine, FEFU, 8 Sukhanova str, Vladivostok, Primorsky region, 690950, Russian Federation
| | - Valeriia Gulaia
- Centre for Genomic and Regenerative Medicine, School of Biomedicine, FEFU, 8 Sukhanova str, Vladivostok, Primorsky region, 690950, Russian Federation
| | - Valeriia Vikhareva
- Laboratory of Pharmacology and Bioassays, School of Biomedicine, FEFU, 8 Sukhanova str, Vladivostok, Primorsky region, 690950, Russian Federation
| | - Boris Zorin
- Microalgal Biotechnology Laboratory, The French Associates Institute for Agriculture and Biotechnology for Drylands, The J. Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede-Boqer Campus, Midreshet Ben-Gurion 8499000, Israel
| | - Alexandra Kalitnik
- Laboratory of Pharmacology and Bioassays, School of Biomedicine, FEFU, 8 Sukhanova str, Vladivostok, Primorsky region, 690950, Russian Federation
| | - Alexander Kagansky
- Centre for Genomic and Regenerative Medicine, School of Biomedicine, FEFU, 8 Sukhanova str, Vladivostok, Primorsky region, 690950, Russian Federation
| |
Collapse
|
22
|
Kunzmann AT, McMenamin ÚC, Spence AD, Gray RT, Murray LJ, Turkington RC, Coleman HG. Blood biomarkers for early diagnosis of oesophageal cancer: a systematic review. Eur J Gastroenterol Hepatol 2018; 30:263-273. [PMID: 29189391 DOI: 10.1097/meg.0000000000001029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Oesophageal cancer prognosis remains poor owing to the inability to detect the disease at an early stage. Nontissue (serum, urinary or salivary) biomarkers potentially offer less invasive methods to aid early detection of oesophageal cancer. We aimed to systematically review studies assessing the relationship between nontissue biomarkers and subsequent development of oesophageal cancer. METHODS Using terms for biomarkers and oesophageal cancer, Medline, EMBASE and Web of Science were systematically searched for longitudinal studies, published until April 2016, which assessed the association between nontissue biomarkers and subsequent oesophageal cancer risk. Random effects meta-analyses were used to calculate pooled relative risk (RR) and 95% confidence intervals (CIs), where possible. RESULTS A total of 39 studies were included. Lower serum pepsinogen I concentrations were associated with an increased risk of oesophageal squamous cell carcinoma (n=3 studies, pooled RR=2.20, 95% CI: 1.31-3.70). However, the association for the pepsinogen I : II ratio was not statistically significant (n=3 studies, pooled RR=2.22, 95% CI: 0.77-6.40), with a large degree of heterogeneity observed (I=68.0%). Higher serum glucose concentrations were associated with a modestly increased risk of total oesophageal cancer (n=3 studies, pooled RR=1.27, 95% CI: 1.02-1.57). No association was observed for total cholesterol and total oesophageal cancer risk (n=3 studies, pooled RR=0.95, 95% CI: 0.58-1.54). Very few studies have assessed other biomarkers for meta-analyses. CONCLUSION Serum pepsinogen I concentrations could aid early detection of oesophageal squamous cell carcinoma. More prospective studies are needed to determine the use of other nontissue biomarkers in the early detection of oesophageal cancer.
Collapse
Affiliation(s)
- Andrew T Kunzmann
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health
| | - Úna C McMenamin
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health
| | - Andrew D Spence
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health
| | - Ronan T Gray
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health
| | - Liam J Murray
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health
| | - Richard C Turkington
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Helen G Coleman
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health
| |
Collapse
|
23
|
Response to TNF-α Is Increasing Along with the Progression in Barrett's Esophagus. Dig Dis Sci 2017; 62:3391-3401. [PMID: 29086334 DOI: 10.1007/s10620-017-4821-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/20/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS Barrett's esophagus, a metaplasia resulting from a long-standing reflux disease, and its progression to esophageal adenocarcinoma (EAC) are characterized by activation of pro-inflammatory pathways, induced by cytokines. METHODS An in vitro cell culture system representing the sequence of squamous epithelium (EPC1 and EPC2), Barrett's metaplasia (CP-A), dysplasia (CP-B) to EAC (OE33 and OE19) was used to investigate TNF-α-mediated induction of interleukin-8 (IL-8). RESULTS IL-6 and IL-8 expressions are increasing with the progression of Barrett's esophagus, with the highest expression of both cytokines in the dysplastic cell line CP-B. IL-8 expression in EAC cells was approx. 4.4-fold (OE33) and eightfold (OE19) higher in EAC cells than in squamous epithelium cells (EPC1 and EPC2). The pro-inflammatory cytokine TNF-α increased IL-8 expression in a time-, concentration-, and stage-specific manner. Furthermore, TNF-α changed the EMT marker profile in OE33 cells by decreasing the epithelial marker E-cadherin and increasing the mesenchymal marker vimentin. The anti-inflammatory compound curcumin was able to repress proliferation and to activate apoptosis in both EAC cell lines. CONCLUSION The increased basal expression levels of IL-8 with the progression of Barrett's esophagus constrain NFκB activation and its contribution in the manifestation of Barrett's esophagus. An anti-inflammatory compound, such as curcumin, could create an anti-inflammatory microenvironment and thus potentially support an increase chemosensitivity in EAC cells.
Collapse
|
24
|
Agoston AT, Srivastava A, Zheng Y, Bueno R, Odze RD, Szallasi Z. Paraneoplastic thrombocytosis is associated with increased mortality and increased rate of lymph node metastasis in oesophageal adenocarcinoma. Pathology 2017; 49:471-475. [PMID: 28668174 DOI: 10.1016/j.pathol.2017.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/15/2017] [Accepted: 04/02/2017] [Indexed: 10/19/2022]
Abstract
Paraneoplastic thrombocytosis has been associated with adverse outcomes in several cancers, but has not been described in oesophageal adenocarcinoma. The aim of our study was to examine the prognostic value of platelet counts in patients with oesophageal adenocarcinoma. A cohort of 584 patients who underwent oesophagectomy for oesophageal adenocarcinoma was identified. Platelet counts, history of neoadjuvant chemoradiation, and clinicopathological factors such as T and N stage, and overall survival were recorded. Patients with elevated platelet count (>450,000/μL) had a higher mortality rate than patients with normal platelet count (150,000-450,000/μL) (hazard ratio = 2.60, p = 0.0013). This effect was seen in patients with and without neoadjuvant chemoradiation therapy. Paraneoplastic thrombocytosis was also associated with increased likelihood of lymph node metastasis compared to normal platelet count (69% versus 31%, p < 0.01). Paraneoplastic thrombocytosis is associated with increased rate of lymph node metastasis and mortality in oesophageal adenocarcinoma. Further studies are needed to examine the mechanisms behind this phenomenon.
Collapse
Affiliation(s)
- Agoston T Agoston
- Brigham and Women's Hospital, Harvard Medical School, Boston, United States.
| | - Amitabh Srivastava
- Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Yifan Zheng
- Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Raphael Bueno
- Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Robert D Odze
- Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Zoltan Szallasi
- Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Young MRI. Redirecting the focus of cancer immunotherapy to premalignant conditions. Cancer Lett 2017; 391:83-88. [PMID: 28130162 PMCID: PMC5925415 DOI: 10.1016/j.canlet.2017.01.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/18/2022]
Abstract
Much progress has been made in introducing immunological treatment approaches for cancer, with lessons learned from both the successes and failures of immunotherapy. Among the challenges of immunotherapeutic approaches for cancer are the multitudes of mechanisms by which cancers are known to subvert the immune defenses. This has led to the incorporation into the immunotherapeutic arsenal strategies by which to overcome the cancer’s immunological blockades. What has been only superficially explored is the immunological milieu of premalignant lesions and the possibility of immunological approaches for the treatment of premalignant lesions so as to prevent secondary premalignant lesions and their progression to cancer. This review discusses the immunological environment associated with premalignant lesions, and the possible missed opportunity of utilizing immunological treatment strategies in the less hostile environment of premalignant lesions as compared to the immune subversive cancer environment.
Collapse
Affiliation(s)
- M Rita I Young
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA; Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
26
|
Cancer Recurrence After Esophagectomy: Impact of Postoperative Infection in Propensity-Matched Cohorts. Ann Thorac Surg 2016; 102:1638-1646. [PMID: 27353482 DOI: 10.1016/j.athoracsur.2016.04.097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/25/2016] [Accepted: 04/28/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Postoperative infection increases cancer recurrence and worsens survival in colorectal cancer, but the relationship for esophagogastric adenocarcinoma after esophagectomy is not well defined. We aimed to determine whether recurrence and survival after minimally invasive esophagectomy for esophagogastric adenocarcinoma were influenced by postoperative infection using propensity-matched analysis. METHODS We abstracted data for 810 patients (1997-2010) and defined exposure as at least 1 in-hospital or 30-day infectious complication (n = 206 [25%]). Using 29 pretreatment/intraoperative variables, patients were propensity-score matched (caliper = 0.05). Time to cancer recurrence and survival (Kaplan-Meier curves and the Breslow test), and associated factors (Cox regression with shared frailty) were assessed. RESULTS After propensity matching (n = 167 pairs), median bias across propensity-score variables was reduced from 12.9% (p < 0.001) to 4.4% (p = 1.000). Postoperative infection was not associated with rate (n = 60 versus 63; McNemar p = 0.736) or time to recurrence in those in whom disease recurred (median, 10.7 versus 11.1 months; Wilcoxon signed-rank p = 0.455) but was associated with shorter overall survival (n = 124 versus 102 deaths; median, 26 versus 41 months; Breslow p = 0.002). After adjusting for age, body mass index, neoadjuvant therapy, sex, comorbidity score, positive resection margins, pathologic stage, R0 resection, and recurrence, postoperative infection was associated with a 44% greater hazard for death (hazard ratio, 1.44; 95% confidence interval, 1.10-1.89). CONCLUSIONS In patients with esophagogastric adenocarcinoma, infections after esophagectomy were not associated with an increased rate or earlier time to recurrence when baseline characteristics associated with infection risk were balanced using propensity-score matching. Despite this, overall survival was shorter in patients with infectious complications. After adjusting for other important survival predictors, infections after esophagectomy continued to be independently associated with worse survival.
Collapse
|
27
|
The tumor microenvironment in esophageal cancer. Oncogene 2016; 35:5337-5349. [PMID: 26923327 PMCID: PMC5003768 DOI: 10.1038/onc.2016.34] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 02/08/2023]
Abstract
Esophageal cancer is a deadly disease, ranking sixth among all cancers in mortality. Despite incremental advances in diagnostics and therapeutics, esophageal cancer still carries a poor prognosis, and thus there remains a need to elucidate the molecular mechanisms underlying this disease. There is accumulating evidence that a comprehensive understanding of the molecular composition of esophageal cancer requires attention to not only tumor cells but also the tumor microenvironment, which contains diverse cell populations, signaling factors, and structural molecules that interact with tumor cells and support all stages of tumorigenesis. In esophageal cancer, environmental exposures can trigger chronic inflammation, which leads to constitutive activation of pro-inflammatory signaling pathways that promote survival and proliferation. Anti-tumor immunity is attenuated by cell populations such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), as well as immune checkpoints like programmed death-1 (PD-1). Other immune cells such as tumor-associated macrophages can have other pro-tumorigenic functions, including the induction of angiogenesis and tumor cell invasion. Cancer-associated fibroblasts secrete growth factors and alter the extracellular matrix (ECM) to create a tumor niche and enhance tumor cell migration and metastasis. Further study of how these TME components relate to the different stages of tumor progression in each esophageal cancer subtype will lead to development of novel and specific TME-targeting therapeutic strategies, which offer considerable potential especially in the setting of combination therapy.
Collapse
|
28
|
Pusung M, Zeki S, Fitzgerald R. Genomics of Esophageal Cancer and Biomarkers for Early Detection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 908:237-63. [PMID: 27573775 DOI: 10.1007/978-3-319-41388-4_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In-depth molecular characterization of esophageal oncogenesis has improved over the recent years. Advancement in molecular biology and bioinformatics has led to better understanding of its genomic landscape. More specifically, analysis of its pathogenesis at the genetic level has uncovered the involvement of a number of tumor suppressor genes, cell cycle regulators, and receptor tyrosine kinases. Due to its poor prognosis, the development of clinically applicable biomarkers for diagnosis, progression, and treatment has been the focus of many research studies concentrating on upper gastrointestinal malignancies. As in other cancers, early detection and subsequent intervention of the preneoplastic condition significantly improves patient outcomes. Currently, clinically approved surveillance practices heavily depend on expensive, invasive, and sampling-error-prone endoscopic procedures. There is, therefore, a great demand to establish clearly reliable biomarkers that could identify those patients at higher risk of neoplastic progression and hence would greatly benefit from further monitoring and/or intervention. This chapter will present the most recent advances in the analysis of the esophageal cancer genome serving as basis for biomarker development.
Collapse
Affiliation(s)
- Mark Pusung
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
29
|
Abstract
The incidence of esophageal adenocarcinoma (EAC), a debilitating and highly lethal malignancy, has risen dramatically over the past 40 years in the United States and other Western countries. To reverse this trend, EAC prevention and early detection efforts by clinicians, academic researchers and endoscope manufacturers have targeted Barrett's esophagus (BE), the widely accepted EAC precursor lesion. Data from surgical, endoscopic and pre-clinical investigations strongly support the malignant potential of BE. For patients with BE, the risk of developing EAC has been estimated at 11- to 125-fold greater than that of the individual at average risk. Nevertheless, screening for BE in symptomatic patients (ie, with symptoms of reflux) and surveillance in patients diagnosed with BE have not had a substantial impact on the incidence, morbidity or mortality of EAC; the overwhelming majority of EAC patients are diagnosed without a pre-operative diagnosis of BE. This article will discuss the current state of the science of esophageal adenocarcinoma prevention, including ideas about carcinogenesis and its underlying genomic and molecular level mechanisms, and suggest strategies for a systems approach to targeted preventive management.
Collapse
Affiliation(s)
- Ellen Richmond
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA.
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
30
|
Shah AK, Cao KAL, Choi E, Chen D, Gautier B, Nancarrow D, Whiteman DC, Saunders NA, Barbour AP, Joshi V, Hill MM. Serum Glycoprotein Biomarker Discovery and Qualification Pipeline Reveals Novel Diagnostic Biomarker Candidates for Esophageal Adenocarcinoma. Mol Cell Proteomics 2015; 14:3023-39. [PMID: 26404905 DOI: 10.1074/mcp.m115.050922] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Indexed: 12/31/2022] Open
Abstract
We report an integrated pipeline for efficient serum glycoprotein biomarker candidate discovery and qualification that may be used to facilitate cancer diagnosis and management. The discovery phase used semi-automated lectin magnetic bead array (LeMBA)-coupled tandem mass spectrometry with a dedicated data-housing and analysis pipeline; GlycoSelector (http://glycoselector.di.uq.edu.au). The qualification phase used lectin magnetic bead array-multiple reaction monitoring-mass spectrometry incorporating an interactive web-interface, Shiny mixOmics (http://mixomics-projects.di.uq.edu.au/Shiny), for univariate and multivariate statistical analysis. Relative quantitation was performed by referencing to a spiked-in glycoprotein, chicken ovalbumin. We applied this workflow to identify diagnostic biomarkers for esophageal adenocarcinoma (EAC), a life threatening malignancy with poor prognosis in the advanced setting. EAC develops from metaplastic condition Barrett's esophagus (BE). Currently diagnosis and monitoring of at-risk patients is through endoscopy and biopsy, which is expensive and requires hospital admission. Hence there is a clinical need for a noninvasive diagnostic biomarker of EAC. In total 89 patient samples from healthy controls, and patients with BE or EAC were screened in discovery and qualification stages. Of the 246 glycoforms measured in the qualification stage, 40 glycoforms (as measured by lectin affinity) qualified as candidate serum markers. The top candidate for distinguishing healthy from BE patients' group was Narcissus pseudonarcissus lectin (NPL)-reactive Apolipoprotein B-100 (p value = 0.0231; AUROC = 0.71); BE versus EAC, Aleuria aurantia lectin (AAL)-reactive complement component C9 (p value = 0.0001; AUROC = 0.85); healthy versus EAC, Erythroagglutinin Phaseolus vulgaris (EPHA)-reactive gelsolin (p value = 0.0014; AUROC = 0.80). A panel of 8 glycoforms showed an improved AUROC of 0.94 to discriminate EAC from BE. Two biomarker candidates were independently verified by lectin magnetic bead array-immunoblotting, confirming the validity of the relative quantitation approach. Thus, we have identified candidate biomarkers, which, following large-scale clinical evaluation, can be developed into diagnostic blood tests. A key feature of the pipeline is the potential for rapid translation of the candidate biomarkers to lectin-immunoassays.
Collapse
Affiliation(s)
- Alok K Shah
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kim-Anh Lê Cao
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Eunju Choi
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia; §School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - David Chen
- ¶School of Information and Communication Technology, Griffith University, Brisbane, Queensland, Australia
| | - Benoît Gautier
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Derek Nancarrow
- ‖QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David C Whiteman
- ‖QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nicholas A Saunders
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Andrew P Barbour
- **School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Virendra Joshi
- ‡‡Ochsner Health System, Gastroenterology, New Orleans, Louisiana
| | - Michelle M Hill
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia;
| |
Collapse
|
31
|
Hardikar S, Song X, Risques RA, Montine TJ, Duggan C, Blount PL, Reid BJ, Anderson GL, Kratz M, White E, Vaughan TL. Obesity and inflammation markers in relation to leukocyte telomere length in a cross-sectional study of persons with Barrett's esophagus. BMC OBESITY 2015; 2:32. [PMID: 26380096 PMCID: PMC4566310 DOI: 10.1186/s40608-015-0063-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 08/20/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Telomere shortening is associated with increasing age, male gender and lifestyle factors such as obesity and smoking. Inflammation has also been implicated in cellular senescence and may promote telomere shortening in chronic conditions such as obesity and diabetes. However, little is known about the relationship between markers of obesity and inflammation, and leukocyte telomere length (LTL). METHODS LTL was measured using quantitative polymerase chain reaction in peripheral leukocytes from 295 individuals diagnosed with Barrett's esophagus (BE) between 1995 and 2009. Data on lifestyle variables including obesity and smoking were collected at in-person interviews. Biomarkers of obesity (leptin, adiponectin), diabetes (glucose, insulin), inflammation (C-reactive protein, Interleukin-6, surface tumor necrosis factor receptor (sTNFR) I & II) and oxidative stress (F2-isoprostanes) were measured in stored blood samples. We examined associations between these covariates and LTL in a cross-sectional analysis using linear and logistic regression models, adjusting for possible confounders. RESULTS LTL was significantly associated with age (r = -0.30, p < 0.001), gender (r = 0.14 for females, p = 0.01) and inversely associated with cigarette pack-years (r = -0.11, p = 0.04). Odds of having short LTL were significantly higher for participants in the highest tertile for sTNF-RI (Odds ratio adjusted for age, gender, smoking, and obesity = 2.19; 95 % CI 1.00-4.85, p-trend = 0.02). LTL was not significantly associated with any other lifestyle factors, including smoking or obesity, or other inflammation-, obesity-/diabetes-related biomarkers measured. CONCLUSIONS Increasing age, male gender, smoking history, and sTNF-RI levels were associated with short LTL among persons with BE but no correlations were observed between LTL and other inflammatory markers or measures of obesity. Larger longitudinal studies are necessary in order to further establish the potential relationships between obesity, inflammation markers and LTL.
Collapse
Affiliation(s)
- Sheetal Hardikar
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, University of Washington, Seattle, WA USA ; 1100 Fairview Ave. N., M4-B402, PO Box 19024, Seattle, WA 98109-1024 USA
| | - Xiaoling Song
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Rosa Ana Risques
- Department of Pathology, University of Washington, Seattle, WA USA
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, WA USA
| | - Catherine Duggan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Patricia L Blount
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Medicine, University of Washington, Seattle, WA USA
| | - Brian J Reid
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Medicine, University of Washington, Seattle, WA USA ; Department of Genome Sciences, University of Washington, Seattle, WA USA
| | - Garnet L Anderson
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Biostatistics, University of Washington, Seattle, WA USA
| | - Mario Kratz
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, University of Washington, Seattle, WA USA ; Department of Genome Sciences, University of Washington, Seattle, WA USA
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, University of Washington, Seattle, WA USA
| | - Thomas L Vaughan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, University of Washington, Seattle, WA USA
| |
Collapse
|
32
|
Abstract
There has been a substantial increase in the incidence of esophageal adenocarcinoma over the past 40 years. Meta-analyses of large prospective cohorts and population-based case-control studies demonstrate consistent associations between obesity and the development of adenocarcinoma of the esophagus and esophago-gastric junction, with an approximate doubling of risk of esophageal adenocarcinoma among patients who are obese, and an almost five-fold increased risk among those with BMI >40 kg/m2. The pathologic precursor, specialized intestinal metaplasia in Barrett's esophagus, is also associated with increased adiposity. Epidemiologic evidence suggests that this cancer risk is not solely due to increased gastro-esophageal reflux, and that adipose tissue itself, in particular visceral adipose, may fuel carcinogenesis through the production of adipokines, cytokines, growth factors, and increased inflammation. The robust epidemiologic evidence linking obesity with esophageal adenocarcinoma makes it an exemplar model for investigating the molecular mechanisms underpinning obesity-associated malignant progression, which are discussed in this review.
Collapse
Affiliation(s)
- Jessie A Elliott
- a 1 Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin & St. James' Hospital, Dublin 8, Ireland
- b 2 Diabetes Complications Research Centre, Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Dublin 4, Ireland
| | - Claire L Donohoe
- a 1 Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin & St. James' Hospital, Dublin 8, Ireland
| | - John V Reynolds
- a 1 Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin & St. James' Hospital, Dublin 8, Ireland
| |
Collapse
|
33
|
Abstract
The incidence of oesophageal adenocarcinoma has risen rapidly over the past four decades. Unfortunately, treatments have not kept pace; unless their cancer is identified at a very early stage, most patients will not survive a year after diagnosis. The beginnings of this widespread problem were first recognized over 25 years ago, yet rates have continued to rise against a backdrop of much improved understanding and management of oesophageal adenocarcinoma. We estimate that only ∼7% of the 10,000 cases of oesophageal adenocarcinoma diagnosed annually in the USA are identified through current approaches to cancer control, and trace pathways by which the remaining 93% are 'lost'. On the basis of emerging data on aetiology and predictive factors, together with new diagnostic tools, we suggest a five-tier strategy for prevention and control that begins with a wide population base and triages individuals into progressively higher risk strata, each with risk-appropriate prevention, screening and treatment options.
Collapse
Affiliation(s)
- Thomas L. Vaughan
- Program in Cancer Epidemiology, Fred Hutchinson Cancer Research Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Rebecca C. Fitzgerald
- Medical Research Council (MRC) Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|