gastrointestinal cancer

胃肠道癌
  • 文章类型: Journal Article
    背景:胰高血糖素样肽-1受体激动剂(GLP-1RA)通常用于降低葡萄糖和减轻体重。然而,它们与胃肠道癌的关系仍不确定.这项荟萃分析评估了使用GLP-1RA治疗的患者的胃肠道癌症风险。
    方法:我们搜索了Medline/PubMed,Embase,和Scopus数据库从开始到2023年11月15日,用于随机对照试验(RCT),并进行至少24周的安全性随访。使用固定效应和随机效应模型计算集合风险比(RR)。使用修订后的Cochrane偏差风险工具评估偏差风险,证据的确定性是使用建议分级评估来确定的,发展,和评估(等级)框架。
    结果:我们纳入了90项RCT,其中有124,791名参与者,每位参与者平均随访3.1年。GLP-1RAs与任何胃肠道癌症的风险之间没有发现显着关联(RR随机=0.99,95%CI:0.86-1.13),或特定部位的胃肠道癌症,包括胆道(RR=0.98,0.54-1.78),结直肠(RR=1.13,0.92-1.39),胆囊(RR=1.32,0.43-4.00),胃(RR=0.88,0.58-1.33),肝(RR=0.79,0.51-1.21),食管(RR=0.70,0.38-1.28),胰腺(RR=1.05,0.77-1.43),和小肠癌(RR=0.78,0.20-3.04)。相应的绝对风险差异排除了对风险的重要影响。其他分析,仅限于安慰剂对照试验,高剂量研究,或随访时间≥5年的人,证实了这些发现。所有结果的偏倚风险普遍较低,证据的确定性较高。
    结论:这项荟萃分析发现GLP-1RA对胃肠道癌症风险没有显著影响。这些药物的长期安全监测仍然很重要。
    背景:CRD42023476762.
    BACKGROUND: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are commonly used for glucose lowering and weight-loss. However, their association with gastrointestinal cancer remains uncertain. This meta-analysis assesses the risk of gastrointestinal cancer in patients treated with GLP-1 RAs.
    METHODS: We searched Medline/PubMed, Embase, and Scopus databases from inception to November 15, 2023, for randomized controlled trials (RCTs) with at least 24 weeks of safety follow-up. Pooled risk ratios (RRs) were calculated using fixed- and random-effect models. Risk of bias was assessed using the revised Cochrane risk-of-bias tool, and certainty of evidence was determined using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) framework.
    RESULTS: We included 90 RCTs with 124,791 participants, with an average follow-up of 3.1 years per participant. No significant association was found between GLP-1 RAs and the risk of any gastrointestinal cancer (RRrandom=0.99, 95% CI: 0.86-1.13), or site-specific gastrointestinal cancers including biliary tract (RR=0.98, 0.54-1.78), colorectal (RR=1.13, 0.92-1.39), gallbladder (RR=1.32, 0.43-4.00), gastric (RR=0.88, 0.58-1.33), hepatic (RR=0.79, 0.51-1.21), oesophageal (RR=0.70, 0.38-1.28), pancreatic (RR=1.05, 0.77-1.43), and small intestine cancer (RR=0.78, 0.20-3.04). The corresponding absolute risk differences excluded important impacts on risk. Additional analyses, limited to placebo-controlled trials, high-dose studies, or those with a follow-up duration of ≥5 years, confirmed these findings. Risk of bias was generally low and the certainty of evidence was high for all outcomes.
    CONCLUSIONS: This meta-analysis found no significant impact of GLP-1 RAs on gastrointestinal cancer risk. Long-term safety monitoring of these agents remains important.
    BACKGROUND: CRD42023476762.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    正在开发许多基于基因组的早期检测筛选测试。这些测试有可能彻底改变当前的单器官筛查模式,尤其是胃肠道癌症。在这次审查中,我们强调了基于前瞻性临床试验的这些基于基因组的早期检测试验的性能.此外,我们讨论了胃肠病学家在诊断评估癌症信号阳性个体方面的专业进步。
    Numerous genomic-based early detection screening tests are being developed. These tests have the potential to revolutionize current single-organ screening paradigms, especially in gastrointestinal cancers. In this review, we underscore the performance of these genomic-based early detection tests based on prospective clinical trials. Moreover, we discuss a professional advancement for gastroenterologists in the diagnostic assessment of individuals who are cancer signal positive.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:老年人在化疗期间有不良反应的风险,包括恶心和疲劳,但许多人也患有头晕和周围神经病变。这可能会导致平衡和步行障碍,并增加跌倒的风险,并影响与健康相关的生活质量。此外,这些症状往往被低估,卫生专业人员的认识不足,导致缺乏对有针对性的评估和康复的关注.我们的目的是检查头晕的患病率,行走平衡受损,以及接受化疗的≥65岁的胃肠道癌症患者的神经病变和跌倒以及这些症状之间的关联。Further,我们旨在检查向肿瘤科医生报告这些症状的患者数量。
    方法:这是一项对≥65岁的胃肠道癌症患者进行的横断面研究,这些患者已经完成了三个或更多个系列的化疗。头晕的患病率,行走平衡受损,神经病,并通过结构化问卷检查这些不良反应的报告。
    结果:在200名患者中(57%为男性,平均年龄74.4岁),头晕的患病率为54%,行走平衡受损的患者的患病率为48%。32%的患者出现神经病变的症状,11%的患者在化疗期间跌倒。神经病变的症状与头晕相关:比值比(OR)1.98(95%置信区间[CI]:1.06;3.71)和平衡受损:OR3.61(95%CI:1.87;6.96)。不到一半的患者(48%)告诉肿瘤学家这些症状。
    结论:在老年癌症患者中,化疗期间头晕和行走平衡受损被低估,但症状严重。应在老年患者化疗期间系统地评估头晕和平衡受损。
    BACKGROUND: Older adults are at risk of adverse effects during chemotherapy including nausea and fatigue, but many also suffer from dizziness and peripheral neuropathy. This may lead to balance and walking impairments and increased risk of falls and affect health-related quality of life. Moreover, these symptoms are often underreported with inadequate awareness among health professionals leading to deficient focus on the need for targeted assessment and rehabilitation. We aimed to examine the prevalence of dizziness, impaired walking balance, and neuropathy and falls in older adults ≥65 years with gastrointestinal cancer receiving chemotherapy and the associations between these symptoms. Further, we aimed to examine the quantity of patients reporting these symptoms to the oncologist.
    METHODS: This is a cross-sectional study among patients ≥65 years with gastrointestinal cancers who have completed three or more series of chemotherapy. The prevalence of dizziness, impaired walking balance, neuropathy, and reporting of these adverse effects was examined through structured questionnaires.
    RESULTS: Of two hundred patients (57 % male, mean age 74.4 years) the prevalence of dizziness was 54 % and the prevalence of patients experiencing impaired walking balance was 48 %. Symptoms of neuropathy was present in 32 % of patients and 11 % experienced falls during chemotherapy. Symptoms of neuropathy was associated with experiencing dizziness: odds ratio (OR) 1.98 (95 % confidence interval [CI]: 1.06; 3.71) and impaired balance: OR 3.61 (95 % CI: 1.87; 6.96). Less than half the patients (48 %) told the oncologist about these symptoms.
    CONCLUSIONS: Dizziness and impaired walking balance during chemotherapy are underreported yet profound symptoms among older patients with cancer. Dizziness and impaired balance should be systematically assessed during chemotherapy among older patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:侵袭性恶性肿瘤,比如胰腺癌,越来越影响年轻人,女性人口。我们的调查集中在观察到的癌症发病率趋势是胰腺癌特有的还是表明各种癌症类型的更广泛的趋势。为了更深入地研究这种现象,我们分析了不同年龄和性别组的癌症发病率趋势.此外,我们探讨了年龄在18~26岁和27~34岁的特定年轻亚组中癌症发病率的差异,以更好地了解年轻个体中出现的发病率趋势.
    方法:本研究从其中一项监测中收集了癌症发病率数据,流行病学,和最终结果癌症登记数据库(SEER22),从2000年到2020年,共有10,183,928例病例。通过Joinpoint趋势分析方法对数据进行分析,以评估癌症发病率的性别和年龄特异性趋势。暴露率报告为平均年百分比变化(AAPC)。
    结果:分析揭示了显著的年龄和性别差异,特别是在18-26岁和27-34岁的人群中。18-26岁女性的胰腺癌发病率增加更多(AAPC,9.37%[95%CI,7.36-11.41%];p<.0001)高于男性(4.43%[95%CI,2.36-6.53%];p<.0001)。值得注意的是,在性别中,年龄,和其他恶性肿瘤,年轻女性患胰腺癌的AAPC最高.此外,胃癌的发病率,骨髓瘤,与男性相比,年轻女性的结直肠恶性肿瘤也显示出更高的AAPC。
    结论:认识到新出现的高致死性恶性肿瘤风险人群对于早期发现和有效的疾病管理至关重要。
    BACKGROUND: Aggressive malignancies, such as pancreatic cancer, are increasingly impacting young, female populations. Our investigation centered on whether the observed trends in cancer incidence were unique to pancreatic cancer or indicative of a broader trend across various cancer types. To delve deeper into this phenomenon, we analyzed cancer incidence trends across different age and sex groups. Furthermore, we explored differences in cancer incidence within specific young subgroups aged 18 to 26 and 27 to 34, to better understand the emerging incidence trend among young individuals.
    METHODS: This study collected cancer incidence data from one of the Surveillance, Epidemiology, and End Results cancer registry databases (SEER22), with 10,183,928 total cases from 2000 to 2020. Data were analyzed through Joinpoint trend analysis approach to evaluate sex- and age-specific trends in cancer incidence. Exposure rates were reported as Average Annual Percentage Changes (AAPCs).
    RESULTS: The analysis revealed significant age and sex-specific disparities, particularly among individuals aged 18-26 and 27-34. Pancreatic cancer incidence rates increased more in females aged 18-26 (AAPC, 9.37% [95% CI, 7.36-11.41%]; p < .0001) than in males (4.43% [95% CI, 2.36-6.53%]; p < .0001). Notably, among gender, age, and other malignancies, young females had the highest AAPCs for pancreatic cancer. Additionally, the incidence of gastric cancer, myeloma, and colorectal malignancies also showed higher AAPCs in young females compared to males.
    CONCLUSIONS: Recognizing emerging risk populations for highly lethal malignancies is crucial for early detection and effective disease management.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:手术切除是胃肠道(GI)癌症的主要治疗方法,但术后骨骼肌丢失(SML)很常见,并与不良预后相关。这项研究旨在确定肌肉变化的模式,检查其与生活质量(QoL)的关系,并探索前3个月SML的预测因子。
    方法:对2021年9月至2022年5月在中国新诊断为胃肠道癌并接受手术的患者进行了前瞻性队列研究。入院时评估骨骼肌质量(SMM)和QoL,7天,1个月,术后3个月.人口统计,临床资料,并收集生物标志物。使用多重插补来估算缺失的数据。使用生长混合物模型分析数据,双变量分析,和逻辑回归。
    结果:共有483名患者完成了基线评估。在242名完成肌肉评估的患者中,92%的人经历过SML。确定了三种不同的肌肉变化模式:57%的术前SMM正常,术后SML轻度,16%术前SMM较低,SML中度,27%的术前肿块正常,但术后SML严重。中度/重度SML与更多的术后并发症相关,健康状况较差,和更高的症状负担。独立预测因素包括高龄,术前肌肉减少症,晚期癌症阶段,预后营养指数低(PNI≤45)。使用估算值时,结果没有变化。
    结论:尽管SML很普遍,肌肉改变的模式在患者之间是异质的。高龄,术前肌肉减少症,晚期癌症阶段,与癌症相关的炎症是中度/重度SML的预测因子,强调早期发现和管理的必要性。
    BACKGROUND: Surgical resection is the primary treatment for gastrointestinal (GI) cancers, but postoperative skeletal muscle loss (SML) is common and linked to poor prognosis. This study aims to identify patterns of muscle change, examine its association with quality of life (QoL), and explore predictors of SML in the first 3 months.
    METHODS: A prospective cohort study was conducted on patients newly diagnosed with GI cancer and undergoing surgery in China between September 2021 and May 2022. Skeletal muscle mass (SMM) and QoL were assessed at admission, 7 days, 1 month, and 3 months post-surgery. Demographic, clinical data, and biomarkers were collected. Missing data were imputed using multiple imputation. Data were analyzed using growth mixture modelling, bivariate analyses, and logistic regression.
    RESULTS: A total of 483 patients completed baseline assessment. Of the 242 patients with complete muscle assessments, 92% experienced SML. Three distinct patterns of muscle change were identified: 57% had normal preoperative SMM with mild postoperative SML, 16% had low preoperative SMM with moderate SML, and 27% had normal preoperative mass but severe postoperative SML. Moderate/severe SML was associated with more postoperative complications, poorer health, and higher symptom burden. Independent predictors included advanced age, preoperative sarcopenia, advanced cancer stage, and low prognostic nutrition index (PNI ≤ 45). The results did not change when using imputed values.
    CONCLUSIONS: Although SML is prevalent, patterns of muscle change are heterogeneous among patients. Advanced age, preoperative sarcopenia, advanced cancer stage, and cancer-related inflammation are predictors for moderate/severe SML, highlighting the need for early detection and management.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Case Reports
    十二指肠腺癌是一种罕见且侵袭性的胃肠道恶性肿瘤,常表现为胃出口梗阻和胆道梗阻等症状。导致延迟诊断和具有挑战性的预后。本病例报告探讨了临床表现,诊断障碍,对一名无明显既往病史的53岁女性晚期十二指肠腺癌进行治疗。患者出现严重的上腹痛,放射到右上腹,恶心,食欲下降。肝酶升高和影像学显示多个肝脏肿块和原发性十二指肠肿块。活检证实中分化腺癌。在分期阶段评估肿瘤标志物,显示明显升高的水平。患者接受了FOLFOX的全身化疗,但面临并发症,包括肺栓塞和神经症状.管理需要多学科方法,整合姑息治疗和支持治疗,以解决症状和改善生活质量。该病例强调了在诊断持续性胃肠道症状时考虑十二指肠腺癌的必要性。它强调了整体治疗方法的必要性,包括量身定制的化疗方案和对并发症的警惕监测。分子谱分析在指导治疗决策方面至关重要,虽然MSI,HER2和PD-1均为阴性,肿瘤没有显示错配修复蛋白缺乏。本文强调早期整合姑息治疗的重要性和综合病理分析在治疗晚期十二指肠腺癌中的价值,为这个复杂的病例提供诊断和治疗策略的见解。
    Duodenal adenocarcinoma is a rare and aggressive gastrointestinal malignancy that frequently presents with symptoms like gastric outlet obstruction and biliary obstruction, leading to delayed diagnosis and challenging prognosis. This case report explores the clinical presentation, diagnostic hurdles, and therapeutic management of late-stage duodenal adenocarcinoma in a 53-year-old woman with no significant prior medical history. The patient presented with severe epigastric pain radiating to the right upper quadrant, nausea, and decreased appetite. Elevated liver enzymes and imaging revealed multiple liver masses and a primary duodenal mass. Biopsies confirmed moderately differentiated adenocarcinoma. Tumor markers were evaluated during the staging phase, showing markedly elevated levels. The patient underwent systemic chemotherapy with FOLFOX but faced complications, including pulmonary emboli and neurological symptoms. Management required a multidisciplinary approach, integrating palliative and supportive care to address symptoms and improve quality of life. The case highlights the necessity of considering duodenal adenocarcinoma when diagnosing persistent gastrointestinal symptoms. It highlights the need for a holistic treatment approach, including tailored chemotherapy regimens and vigilant monitoring of complications. Molecular profiling was crucial in guiding treatment decisions, although MSI, HER2, and PD-1 were negative, and the tumor showed no mismatch repair protein deficiency. This article emphasizes the importance of early integration of palliative care and the value of comprehensive pathological analysis in managing advanced duodenal adenocarcinoma, providing insights into diagnostic and therapeutic strategies for this complex case.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胃癌已成为世界范围内严重关注的健康问题,强调早期诊断措施对改善患者预后至关重要。而传统的组织学图像分析被视为临床的金标准,它是劳动密集型和手动的。认识到这个问题,人们越来越关注使用计算机辅助诊断工具来帮助病理学家进行诊断工作。特别是,深度学习(DL)已成为该领域有前途的解决方案。然而,当前的DL模型在提取大量视觉特征以进行正确分类的能力方面仍然受到限制。为了解决这个限制,这项研究提出了使用集成模型,它融合了几种深度学习架构的功能,并使用许多模型的聚合知识来提高分类性能,允许更准确和有效的胃癌检测。为了确定这些建议的模型的性能,这项研究将它们与其他作品进行了比较,所有这些都是基于胃组织病理学小尺寸图像数据库,一个公开的胃癌数据集。这项研究表明,集成模型在所有子数据库中都实现了很高的检测精度,平均准确率超过99%。具体来说,ResNet50、VGGNet、ResNet34的性能优于EfficientNet和VitNet。对于80×80像素的子数据库,ResNet34的准确度约为93%,VGGNet实现了94%,合奏模型以99%的成绩出色。在120×120像素的子数据库中,集成模型显示99%的准确性,VGGNet97%,和ResNet50约97%。对于160×160像素的子数据库,集成模型再次达到99%的准确率,VGGNet98%,ResNet5098%,和效率网92%,突出显示合奏模型在所有分辨率中的卓越性能。总的来说,集成模型在三个子像素类别中始终提供99%的准确性。这些发现表明,集成模型可以成功地从较小的补丁中检测关键特征并实现高性能。这些发现将帮助病理学家使用组织病理学图像诊断胃癌,导致更早的识别和更高的患者生存率。
    Gastric cancer has become a serious worldwide health concern, emphasizing the crucial importance of early diagnosis measures to improve patient outcomes. While traditional histological image analysis is regarded as the clinical gold standard, it is labour intensive and manual. In recognition of this problem, there has been a rise in interest in the use of computer-aided diagnostic tools to help pathologists with their diagnostic efforts. In particular, deep learning (DL) has emerged as a promising solution in this sector. However, current DL models are still restricted in their ability to extract extensive visual characteristics for correct categorization. To address this limitation, this study proposes the use of ensemble models, which incorporate the capabilities of several deep-learning architectures and use aggregate knowledge of many models to improve classification performance, allowing for more accurate and efficient gastric cancer detection. To determine how well these proposed models performed, this study compared them with other works, all of which were based on the Gastric Histopathology Sub-Size Images Database, a publicly available dataset for gastric cancer. This research demonstrates that the ensemble models achieved a high detection accuracy across all sub-databases, with an average accuracy exceeding 99%. Specifically, ResNet50, VGGNet, and ResNet34 performed better than EfficientNet and VitNet. For the 80 × 80-pixel sub-database, ResNet34 exhibited an accuracy of approximately 93%, VGGNet achieved 94%, and the ensemble model excelled with 99%. In the 120 × 120-pixel sub-database, the ensemble model showed 99% accuracy, VGGNet 97%, and ResNet50 approximately 97%. For the 160 × 160-pixel sub-database, the ensemble model again achieved 99% accuracy, VGGNet 98%, ResNet50 98%, and EfficientNet 92%, highlighting the ensemble model\'s superior performance across all resolutions. Overall, the ensemble model consistently provided an accuracy of 99% across the three sub-pixel categories. These findings show that ensemble models may successfully detect critical characteristics from smaller patches and achieve high performance. The findings will help pathologists diagnose gastric cancer using histopathological images, leading to earlier identification and higher patient survival rates.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胃肠道(GI)癌症给全球带来了巨大的健康负担,强调需要更深入地了解其复杂的发病机制和治疗策略。这篇综述探讨了肿瘤内微生物群之间的相互作用,肿瘤代谢,和主要类型的胃肠道癌症(包括食道癌,胃,肝脏,胰腺,和结肠直肠癌),总结最近的研究,阐明其临床意义和未来的方向。最近的研究揭示了胃肠道肿瘤中微生物特征的改变,影响肿瘤进展,免疫反应,和治疗结果。菌群失调诱导的肿瘤代谢改变,包括糖酵解,脂肪酸代谢,和氨基酸代谢,在癌症进展和治疗抗性中起关键作用。将分子机制和潜在生物标志物整合到这种理解中进一步增强了肿瘤内微生物群组成和靶向微生物群介导的肿瘤代谢的治疗机会的预后意义。尽管取得了进步,在理解肿瘤微环境(TME)内的动态相互作用方面仍然存在挑战。未来的研究方向,包括先进的组学技术和前瞻性临床研究,为胃肠道癌症的精准肿瘤学和个性化治疗干预提供了有希望的途径。总的来说,将基于微生物群的方法和分子生物标志物整合到胃肠道癌症管理中,有望改善患者预后和生存率.
    Gastrointestinal (GI) cancers impose a substantial global health burden, highlighting the necessity for deeper understanding of their intricate pathogenesis and treatment strategies. This review explores the interplay between intratumoral microbiota, tumor metabolism, and major types of GI cancers (including esophageal, gastric, liver, pancreatic, and colorectal cancers), summarizing recent studies and elucidating their clinical implications and future directions. Recent research revealed altered microbial signatures within GI tumors, impacting tumor progression, immune responses, and treatment outcomes. Dysbiosis-induced alterations in tumor metabolism, including glycolysis, fatty acid metabolism, and amino acid metabolism, play critical roles in cancer progression and therapeutic resistance. The integration of molecular mechanisms and potential biomarkers into this understanding further enhances the prognostic significance of intratumoral microbiota composition and therapeutic opportunities targeting microbiota-mediated tumor metabolism. Despite advancements, challenges remain in understanding the dynamic interactions within the tumor microenvironment (TME). Future research directions, including advanced omics technologies and prospective clinical studies, offer promising avenues for precision oncology and personalized treatment interventions in GI cancer. Overall, integrating microbiota-based approaches and molecular biomarkers into GI cancer management holds promise for improving patient outcomes and survival.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胃肠道癌症是全球健康挑战,因为它的患病率急剧增加,并且是癌症相关死亡率的主要原因。越来越多的证据表明肠道微生物衍生的代谢产物在胃肠道癌症进展和治疗中的重要作用。微生物代谢物由利用外在饮食组分和内在宿主产生的化合物的肠道微生物群产生。同时,某些类别的代谢物,如短链脂肪酸,胆汁酸,色氨酸,和吲哚衍生物,与胃肠道恶性肿瘤有关.在这次审查中,微生物代谢产物的主要类别及其对包括结直肠癌在内的各种胃肠道癌症的影响,胃癌,和肝细胞癌,已被介绍。还探索了微生物代谢物作为预测生物标志物在胃肠道癌症早期诊断和预后中的应用。此外,进一步评估了靶向微生物代谢物对抗胃肠道癌的策略的治疗潜力。
    Gastrointestinal cancer is a worldwide health challenge due to its dramatically increasing prevalence and as a leading cause of cancer-related mortality. Increasing evidence has illustrated the vital role of gut microbes-derived metabolites in gastrointestinal cancer progression and treatment. Microbial metabolites are produced by the gut microbiota that utilizes both extrinsic dietary components and intrinsic host-generated compounds. Meanwhile, certain categories of metabolites such as short-chain fatty acids, bile acids, tryptophan, and indole derivatives, are linked to gastrointestinal malignancy. In this review, the major classes of microbial metabolites and their impacts on various gastrointestinal cancers including colorectal cancer, gastric cancer, and hepatocellular carcinoma, have been introduced. The application of microbial metabolites as predictive biomarkers for early diagnosis and prognosis of gastrointestinal cancer has also been explored. In addition, therapeutic potential of strategies that target microbial metabolites against gastrointestinal cancer is further evaluated.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    癌症是全世界死亡的主要原因。全球癌症发病率和死亡率的约三分之一与胃肠道(GI)癌症有关。在过去的几年里,胃肠道肿瘤的患者来源的类器官(PDO)模型得到了快速发展.通过在体外密切模拟其母体肿瘤的分子特性,PDO已经成为个性化医疗和药物发现的强大工具。这里,我们回顾了目前关于PDOs在临床药物治疗策略优化中的应用及其在临床前药物开发中日益重要的文献。我们讨论了胃肠道肿瘤PDO的优点和局限性,并概述了基于微流体的策略,这些策略可以提高PDO模型的吞吐量,从而在个性化医疗和药物发现过程中获得最大的好处。
    Cancer is a leading cause of death worldwide. Around one-third of the total global cancer incidence and mortality are related to gastrointestinal (GI) cancers. Over the past few years, rapid developments have been made in patient-derived organoid (PDO) models for gastrointestinal cancers. By closely mimicking the molecular properties of their parent tumors in vitro, PDOs have emerged as powerful tools in personalized medicine and drug discovery. Here, we review the current literature on the application of PDOs of common gastrointestinal cancers in the optimization of drug treatment strategies in the clinic and their rising importance in pre-clinical drug development. We discuss the advantages and limitations of gastrointestinal cancer PDOs and outline the microfluidics-based strategies that improve the throughput of PDO models in order to extract the maximal benefits in the personalized medicine and drug discovery process.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号