• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 1
  • 1
  • Tagged with
  • 7
  • 7
  • 5
  • 4
  • 3
  • 3
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Clonal interactions in Barrett's carcinogenesis

Zeki, Sebastian Simon January 2013 (has links)
Introduction: Barrett’s oesophagus (BO) is a metaplastic premalignant disease which can undergo a metaplasia-­‐dysplasia-­‐adenocarcinoma pathway. It represents an example of field cancerization by which an area occupied by BO can undergo molecular and genetic changes associated with carcinogenesis without being phenotypically cancerous. Previous work suggested that non-­‐cancerous BO contains a monoclonal population. More recent work demonstrated that premalignant Barrett’s fields are polyclonal suggesting that clonal interactions may be important in carcinogenesis. It is the aim of this thesis to further investigate clonal interactions in BO by understanding the effects of therapy in altering the relationships of clonal populations in BO, by assessing the relationship of clonal populations in dysplasia as compared with the associated cancer, and by attempting to elucidate a potential molecular mechanism of clonal interactions. Results: The overall results can be summarised as follows: 1.Premalignant clonal populations are well mixed allowing for clonal interactions. However, the adenocarcinoma associated with high grade dysplasia is monoclonal and derived from clonal populations found in the dysplasia, indicating possible clonal interactions during carcinogenesis. 2. Patients with persistent disease after endoscopy retain the same clonal populations. However, the clonal populations of recurrent disease changes such that new clonal populations arise or may benefit from the extinction of others. 3. These clonal populations may be derived from deep submucosal glands or may be found in phenotypically normal squamous epithelium indicating a common stem cell origin. 4. A possible mechanism of clonal interaction may be the senescence associated secretory phenotype: senescence is abundant in BO and can cause proliferation in neighbouring cells in vitro. Conclusion: This thesis has investigated the implications of clonal interactions in BO. The demonstration of temporal clonal heterogeneity as a result of endoscopic therapy, as well as spatial clonal heterogeneity possibly resulting in carcinogenesis, asks for a mechanistic explanation of clonal interactions. The consequences of senescence may well provide one such mechanism.
2

On the progression of Barrett's oesophagus to Barrett's adenocarcinoma

Khan, Shabuddin January 2017 (has links)
Barrett's oesophagus (BO) is the major precursor of oesophageal adenocarcinoma (OA) and we do not understand the dynamics of the evolution of BO in order to identify patients at high risk of cancer. Studies have proposed that BO is a monoclonal lesion, however recent work has shown that there are multiple independent clones present. Project 1: Determines the evolution of polyclonal dysplasia through sequencing and mapping clones onto tissue sections. I show that several cases are polyclonal but in each case only one clone progresses to cancer, suggesting oesophageal cancers are monoclonal outgrows from polyclonal Barrett's dysplasia. Project 2: Aims to understand the clonal relationship between cells in glands displaying basal crypt dysplasia-like atypia (BCDA), as it is unclear whether those cells in the upper part of the gland arise from the same stem cell that generates the gland bases. Glands displaying BCDA show a common mutation between the dysplastic base and non-dysplastic surface suggesting a common cell of origin. Project 3: 50% of patients who undergo oesophagectomy for OA develop post-oesophagectomy Barrett's (neo- BO) within 3-5 years possibly due to a field effect, wherein pre-neoplastic cells remain post-resection in histologically normal areas of epithelium predisposing the patient to cancer recurrence. Here I show that no genetic link between the neo-BO and the cancer is present. Immunohistochemical analysis shows that neo- Barrett's glands are gastric in nature. Project 4: The stem cell dynamics and clonal expansion rates of BO are unknown. Here I employed diversity analysis of methylation patterns of CpG islands in the promoter regions of non-expressed genes as a molecular clock. My data suggests that 3-4 stem cells are found in each Barrett's gland. Methylation patterns within a gland were less diverse compared to adjacent and distant glands, suggesting BO is characterized by long periods of stasis followed by bursts of clonal expansions.
3

Characterisation of copy number changes in the progression of Barrett's oesophagus

Gregson, Eleanor January 2018 (has links)
Introduction: The main risk factor for the development of oesophageal adenocarcinoma is Barrett’s oesophagus (BE). To diagnose those patients who will progress to cancer early to improve the dismal survival rate of oesophageal adenocarcinoma, patients with BE undergo regular endoscopic surveillance. The vast majority of patients, however, will never progress and are therefore monitored unnecessarily. Copy number changes have been shown to be important in the progression of BE to oesophageal adenocarcinoma (Li et al., 2014). Shallow whole genome sequencing (sWGS) has been established as a cost-effective method of investigating copy number changes in formalin fixed paraffin embedded (FFPE) tissue (Scheinin et al., 2014). We hypothesised that copy number alterations may be valuable markers in disease progression and aimed to characterise them in the progression of Barrett’s using sWGS in order to predict progression in patients from a point in time as close to baseline endoscopy as possible and to integrate p53 staining. Methods: To optimise sWGS we compared 50X WGS on frozen tissue with 0.1X WGS from FFPE tumour material from the same patient. To address poor cellularity in endoscopic biopsies, shallow WGS data from a 50% cellularity biopsy with a 90% frozen sample from a single patient were compared. Accounting for poor biopsy cellularity 0.4X coverage was used. We performed FFPE shallow WGS on 806 samples from an 89-patient cohort comprising a 1:1 ratio of patients who progressed to high grade dysplasia (HGD) and patients who never progressed. 1-31 samples per patient were collected over time and space throughout surveillance. Non-progressors had significantly longer follow-up (p-value = 0.0008). Data was processed based on published bioinformatic pipelines. Copy number analysis was carried out using a generalised linear model (GLM) in order to develop a predictive algorithm. Results: During optimisation, ˃85% of copy number changes were detected in both frozen and FFPE samples from spatially distinct regions of an individual tumour. We found 91% and 93% agreement in copy number calls using orthogonal platforms between 90% (frozen) and 50% (FFPE) cellularity samples from one tumour. In the 806 sample Barrett’s cohort, we observed larger copy number alterations in patients who progressed to cancer compared with non-progressors and significantly more CN alterations in progressor patients (p-value ˂ 0.001). More cancer-associated genes were affected in progressors and we observed significant heterogeneity between patients. There was also a greater level of complexity seen in the progressor patients when analysed using affinity propagation clustering. These data allowed us to develop a regression model to predict progression. Using the GLM model, we successfully classified samples as early as progressor or not with an AUC of 85.75% and a sensitivity and specificity of 84 and 79% respectively. At the patient level 94% progressor patients had at least one sample classified as at risk of progression and non-dysplastic progressor samples were classified as early as 13 years prior to HGD diagnosis. Depending on the classification threshold used, all samples over time and space were not classified as being at risk of progression in at least 60% patients who have not yet progressed to HGD/cancer. We observed 2 pathways to progression supporting previous observations. 90% of progressors had samples prior to their HGD or cancer diagnosis classified as being predisposed to progression suggestive of genetically unstable lesions from early on in surveillance that progressed to HGD over time. The remaining 10% appeared as non-progressors until their diagnosis of HGD. We investigated p53 expression in our patient cohort as the only biomarker to have successfully transitioned into the clinic for Barrett’s surveillance. Whilst we found our cohort to be representative in staining compared to other published cohorts, it did not contribute to the GLM and the copy number data out-performed the use of p53 IHC in the context of Barrett’s surveillance. Conclusions: We have optimised the use of shallow WGS in oesophageal adenocarcinoma and Barrett’s. Using these copy number data, we can confidently distinguish between patients who will progress to cancer and the majority of patients who will never progress. This approach has led to the development of a model for predicting progression in the clinical setting which is promising for further clinical validation.
4

Chemoprevention in a validated rat model of oesophageal adenocarcinoma

Hindmarsh, Andrew January 2012 (has links)
The UK has experienced an increase in the incidence of oesophageal adenocarcinoma (OAC) in recent years. The prognosis for patients with OAC remains poor with currently available treatments prompting a search for alternative ‘chemopreventive’ treatments that inhibit oesophageal carcinogenesis. Both non-steroidal anti-inflammatory drugs (NSAIDS) and flavonoids are associated with a significant risk reduction for developing OAC in epidemiological studies. The aim of this study was to validate Levrat’s surgical model of OAC in the rat, and assess the chemopreventive effects of the NSAID aspirin, and the flavonoid quercetin on the development of OAC in the validated rat model. METHODS: Levrat’s model was validated in a time course experiment. Morphological and molecular events occurring in the distal oesophagus during disease progression were determined and compared to human disease. The effect of aspirin and quercetin on disease initiation and progression was determined by commencing treatment either before the onset of reflux, or 4-weeks afterwards. The incidence of Barrett’s oesophagus (BO) and OAC within each group was determined, along with methylation levels of the ESR-1, p16 and HPP1 gene promoter regions. RESULTS: The morphological and molecular changes in the distal oesophagus of the rat model are broadly consistent with those reported in human disease. The incidence of OAC was significantly lower in aspirin treated rats. A non-significant reduction in incidence of OAC was observed with quercetin treatment. Timing of treatment with regard to onset of reflux had no significant effect on OAC development in either treatment group. Neither treatment significantly effected methylation levels within the gene promoters examined. CONCLUSION: Use of Levrat’s model as a model of human OAC seems justified. Aspirin inhibits development of oesophageal adenocarcinoma induced by reflux in this rat model. No additional reduction in cancer incidence is observed if treatment is commenced prior to inception of reflux disease.
5

The involvement of bacteria in the progression of Barrett's oesophagus to adenocarcinoma of the oesophagus

Blackett, Katie January 2010 (has links)
Barrett's oesophagus (BO) arises from chronic gastro-oesophageal reflux disease(GORD). Patients have an increased risk of adenocarcinoma (ADC), which is the sixth most common cause of cancer mortality in the UK. All ADC develop from BO, and over the last twenty years there has been a marked increase in both conditions. The reasons for this are not known, however, as with some forms of gastric cancer, it is possible that there may be a bacterial aetiology. This study employed both culturebased and molecular techniques to characterise microbial communities colonising the distal oesophageal mucosae in individuals with GORD, BO and ADC, together with healthy controls. Furthermore, in vitro models were designed to create an oral microbiota, from which an oesophageal community could develop. Microbial analysis identified a shift in oesophageal population composition with disease progression, with an incremental increase in total eubacterial scores related to the metaplasia-dysplasia sequence. Additionally, an increased proportion of Gram negative species and potentially pathogenic organisms, such as Peptostreptococcus were identified. Campylobacter spp. were isolated from 75%, 50% and 60% of GORD, BO and ADC patients, respectively, compared with 20% of controls. Helicobacter pylori, which has been proposed to be protective in oesophageal disease, was significantly reduced in disease, especially in ADC patients. In vitro models were successful, with a simple oral microbiota leading to the development of unique, varied oesophageal populations representative of those found in vivo. Additionally, after exposure of this community to bile acid, population dynamics were altered, with an increase in Gram negative species, associated with a rise in haemolytic and mucinolytic activities. Exposure of oesophageal cell lines to these stressed biofilms resulted in increased cell death, and in some cases, amplified expression of p53 and COX-2. In conclusion, this research proved an association between bacterial composition and oesophageal disease. With progression to adenocarcinoma, the community becomes increasingly diversified and Gram negative in character, and therefore, is proposed to be more pathogenic. Further research is required to investigate causal relationships, through which mechanisms for disease initiation and/or maintenance can be understood.
6

Sunkių gastroezofaginio refliukso formų charakteristika bei Bareto stemplės sindromo progresavimo rizikos veiksniai / Characteristics of severe forms of gastro-oesophageal reflux disease and risk factors of progression of Barrett’s oesophagus

Kriukas, Darius 02 December 2008 (has links)
1. Nustatyti erozinio ezofagito ir jo komplikacijos – ikivėžinio susirgimo, Bareto stemplės, dažnį tarp endoskopiniam tyrimui atsiųstų pacientų, turinčių viršutiniojo virškinamojo trakto skundų ir/ar „pavojaus“ simptomų. 2. Nustatyti klinikinius, endoskopinius ir morfologinius požymius, susijusius su gastroezofaginio reflukso ligos sunkumu. 3. Nustatyti klinikinius, endoskopinius ir morfologinius požymius, susijusius su ikivėžiniu susirgimu – Bareto stemple. 4. Nustatyti skrandžio, skrandžio – stemplės jungties ir stemplės gleivinės morfologinius pakitimus, sergant įvairaus sunkumo GERL ir Bareto stemple. 5. Išanalizuoti trumpo ir ilgo segmento Bareto stemple sergančių asmenų klinikinius, endoskopinius ir morfologinius ypatumus. 6. Nustatyti rizikos veiksnius, įtakojančius ikivėžinio susirgimo - Bareto stemplės - progresavimą dvejų metų stebėjimo laikotarpiu. / 1. To investigate the prevalence of erosive esophagitis and its complication -precancerous disease, Barrett‘s oesophagus, in patients referred to upper diagnostic endoscopy with upper gastrointestinal and/or „alarm“ symptoms. 2. To establish clinical, endoscopic and morphological signs associated with severity of gastro-oesophageal reflux disease. 3. To establish clinical, endoscopic and morphological signs associated with precancerous disease - Barrett‘s oesophagus. 4. To determine morphological alterations of stomach, gastro-oesophageal junction and oesophagus mucosa of different severity of GORD and Barrett‘s oesophagus. 5. To analyse clinical, endoscopic and morphological peculiarities of long and short segments of Barrett’s oesophagus. 6. To investigate risk factors associated with progression of precancerous disease, Barrett‘s oesophagus, during two years follow-up period.
7

Multitrait genetic association analysis identifies 50 new risk loci for gastro-oesophageal reflux, seven new loci for Barrett’s oesophagus and provides insights into clinical heterogeneity in reflux diagnosis

Ong, Jue-Sheng, An, Jiyuan, Law, Matthew H., Nandakumar, Priyanka, Schumacher, Johannes, Gockel, Ines, Bohmer, Anne, Jankowski, Janusz, Palles, Claire, Olsen, Catherine M., Neale, Rachel E., Fitzgerald, Rebecca, Thrift, Aaron P., Vaughan, Thomas L., Buas, Matthew F., Hinds, David A., Gharahkhani, Puya, Kendall, Bradley J., MacGregor, Stuart, ., 23andMe Research Team, ., Esophageal cancer consortium 05 June 2023 (has links)
Objective: Gastro-oesophageal reflux disease (GERD) has heterogeneous aetiology primarily attributable to its symptom-based definitions. GERD genome-wide association studies (GWASs) have shown strong genetic overlaps with established risk factors such as obesity and depression. We hypothesised that the shared genetic architecture between GERD and these risk factors can be leveraged to (1) identify new GERD and Barrett's oesophagus (BE) risk loci and (2) explore potentially heterogeneous pathways leading to GERD and oesophageal complications. Design: We applied multitrait GWAS models combining GERD (78 707 cases; 288 734 controls) and genetically correlated traits including education attainment, depression and body mass index. We also used multitrait analysis to identify BE risk loci. Top hits were replicated in 23andMe (462 753 GERD cases, 24 099 BE cases, 1 484 025 controls). We additionally dissected the GERD loci into obesity-driven and depression-driven subgroups. These subgroups were investigated to determine how they relate to tissue-specific gene expression and to risk of serious oesophageal disease (BE and/or oesophageal adenocarcinoma, EA). Results: We identified 88 loci associated with GERD, with 59 replicating in 23andMe after multiple testing corrections. Our BE analysis identified seven novel loci. Additionally we showed that only the obesity-driven GERD loci (but not the depression-driven loci) were associated with genes enriched in oesophageal tissues and successfully predicted BE/EA. Conclusion: Our multitrait model identified many novel risk loci for GERD and BE. We present strong evidence for a genetic underpinning of disease heterogeneity in GERD and show that GERD loci associated with depressive symptoms are not strong predictors of BE/EA relative to obesity-driven GERD loci.

Page generated in 0.0759 seconds