1
|
Hicheri C, Azimuddin AM, Kortum A, Bailey J, Tang Y, Schwarz RA, Rosen D, Jain S, Mansour NM, Groth S, Vasavada S, Rao A, Maliga A, Gallego L, Carns J, Anandasabapathy S, Richards-Kortum R. Design and Evaluation of ScanCap: A Low-Cost, Reusable Tethered Capsule Endoscope with Blue-Green Illumination Imaging for Unsedated Screening and Early Detection of Barrett's Esophagus. Bioengineering (Basel) 2024; 11:557. [PMID: 38927792 PMCID: PMC11200367 DOI: 10.3390/bioengineering11060557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 06/28/2024] Open
Abstract
Esophageal carcinoma is the sixth-leading cause of cancer death worldwide. A precursor to esophageal adenocarcinoma (EAC) is Barrett's Esophagus (BE). Early-stage diagnosis and treatment of esophageal neoplasia (Barrett's with high-grade dysplasia/intramucosal cancer) increase the five-year survival rate from 10% to 98%. BE is a global challenge; however, current endoscopes for early BE detection are costly and require extensive infrastructure for patient examination and sedation. We describe the design and evaluation of the first prototype of ScanCap, a high-resolution optical endoscopy system with a reusable, low-cost tethered capsule, designed to provide high-definition, blue-green illumination imaging for the early detection of BE in unsedated patients. The tethered capsule (12.8 mm diameter, 35.5 mm length) contains a color camera and rotating mirror and is designed to be swallowed; images are collected as the capsule is retracted manually via the tether. The tether provides electrical power and illumination at wavelengths of 415 nm and 565 nm and transmits data from the camera to a tablet. The ScanCap prototype capsule was used to image the oral mucosa in normal volunteers and ex vivo esophageal resections; images were compared to those obtained using an Olympus CV-180 endoscope. Images of superficial capillaries in intact oral mucosa were clearly visible in ScanCap images. Diagnostically relevant features of BE, including irregular Z-lines, distorted mucosa, and dilated vasculature, were clearly visible in ScanCap images of ex vivo esophageal specimens.
Collapse
Affiliation(s)
- Cheima Hicheri
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Ahad M. Azimuddin
- Houston Methodist Hospital, Houston, TX 77030, USA;
- Texas A&M School of Medicine, Houston, TX 77030, USA
| | - Alex Kortum
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Joseph Bailey
- Rice360 Institute for Global Health Technologies, Rice University, Houston, TX 77030, USA
| | - Yubo Tang
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Richard A. Schwarz
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Daniel Rosen
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | - Shilpa Jain
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | | | - Shawn Groth
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | | | - Ashwin Rao
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | | | - Leslie Gallego
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | - Jennifer Carns
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | | | | |
Collapse
|
2
|
Che J, Zhao Y, Gu B, Li S, Li Y, Pan K, Sun T, Han X, Lv J, Zhang S, Fan B, Li C, Wang C, Wang J, Zhang T. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with the progression of gastroesophageal cancer. BMC Cancer 2023; 23:1238. [PMID: 38102546 PMCID: PMC10724912 DOI: 10.1186/s12885-023-11744-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Previous metabolic studies in upper digestive cancer have mostly been limited to cross-sectional study designs, which hinders the ability to effectively predict outcomes in the early stage of cancer. This study aims to identify key metabolites and metabolic pathways associated with the multistage progression of epithelial cancer and to explore their predictive value for gastroesophageal cancer (GEC) formation and for the early screening of esophageal squamous cell carcinoma (ESCC). METHODS A case-cohort study within the 7-year prospective Esophageal Cancer Screening Cohort of Shandong Province included 77 GEC cases and 77 sub-cohort individuals. Untargeted metabolic analysis was performed in serum samples. Metabolites, with FDR q value < 0.05 and variable importance in projection (VIP) > 1, were selected as differential metabolites to predict GEC formation using Random Forest (RF) models. Subsequently, we evaluated the predictive performance of these differential metabolites for the early screening of ESCC. RESULTS We found a distinct metabolic profile alteration in GEC cases compared to the sub-cohort, and identified eight differential metabolites. Pathway analyses showed dysregulation in D-glutamine and D-glutamate metabolism, nitrogen metabolism, primary bile acid biosynthesis, and steroid hormone biosynthesis in GEC patients. A panel of eight differential metabolites showed good predictive performance for GEC formation, with an area under the receiver operating characteristic curve (AUC) of 0.893 (95% CI = 0.816-0.951). Furthermore, four of the GEC pathological progression-related metabolites were validated in the early screening of ESCC, with an AUC of 0.761 (95% CI = 0.716-0.805). CONCLUSIONS These findings indicated a panel of metabolites might be an alternative approach to predict GEC formation, and therefore have the potential to mitigate the risk of cancer progression at the early stage of GEC.
Collapse
Affiliation(s)
- Jiajing Che
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yongbin Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Bingbing Gu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuting Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yunfei Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Keyu Pan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Tiantian Sun
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xinyue Han
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jiali Lv
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuai Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Bingbing Fan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chunxia Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Cheng Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Jialin Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China.
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
3
|
Mazidimoradi A, Banakar N, Khani Y, Allahqoli L, Salehiniya H. Current status and temporal trend in incidence, death, and burden of esophageal cancer from 1990-2019. Thorac Cancer 2023; 14:2408-2458. [PMID: 37443420 PMCID: PMC10447176 DOI: 10.1111/1759-7714.15028] [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] [Received: 05/09/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Esophageal cancer (EC) is one of the world's most unknown and deadly cancers. This study aimed to provide updated epidemiological indicators and the recent trend of EC by age group, gender, and geographical region in the world. METHODS Annual case data and age-standardized rates (ASRs) of epidemiological indicators of EC were collected from the 2019 Global Burden of Disease (GBD) study from 1990 to 2019 in 204 countries and territories based on the sociodemographic index (SDI). Relative difference (%), average annual percentage change (AAPC), and the male/female ratio were calculated. Data are reported in values and 95% confidence interval (CI). RESULTS EC age-standardized incidence rates (ASIR) decreased by 19%, age-standardized death rates (ASDR) decreased by 25%, and disability-adjusted life-years ASR (DALYs ASR) decreased by 30% from 1990 to 2019. The higher number of EC cases was in men aged 50 to 69 years and in women aged over 70. From 1990 to 2019, Middle SDI countries experienced a decline in the ASIR and ASDR of EC. The High SDI countries had an increasing ASDR trend. In World Bank High-Income countries, the ASIR of EC has remained unchanged and decreased in other regions. The Asia continent has the highest rate of incidence, mortality, and burden of EC and the highest rate of reduction. East Asia, Southern Sub-Saharan Africa, and Eastern Sub-Saharan Africa respectively have the highest ASIR of EC. Central Asia has experienced the greatest decrease in the ASIR and ASDR of EC, the countries of Central Europe had a steady ASIR and High-Income North America had an increasing trend in ASIR and ASDR. The burden of EC shows a decreasing trend worldwide. Central and East Asia regions have the highest rate and the highest increase in the burden of EC. CONCLUSION Based on great variation in the geographical distribution of epidemiological indicators of EC, investigating the reasons for this diversity requires more studies to be conducted in the field of prevention, distribution of risk factors, and implementation of screening methods with high cost-effectiveness, and access to treatment methods. The provision of regional solutions may be more effective than global strategies.
Collapse
Affiliation(s)
| | - Niloofar Banakar
- Student Research CommitteeShiraz University of medical sciencesShirazIran
| | - Yousef Khani
- Clinical Research Development Unit, Shahid Madani HospitalAlborz University of Medical SciencesKarajIran
- School of Public Health and SafetyShahid Beheshti University of Medical SciencesTehranIran
| | - Leila Allahqoli
- Midwifery Department, Ministry of Health and Medical EducationTehranIran
| | - Hamid Salehiniya
- Department of Epidemiology and Biostatistics, School of Health, Social Determinants of Health Research CenterBirjand University of Medical SciencesBirjandIran
| |
Collapse
|
4
|
Xu J, Benson ME, Granlund LM, Gehrke S, Stanfield D, Weiss J, Pfau P, Soni A, Cox BL, Petry G, Swader RA, Reichelderfer M, Li Z, Atrukstang T, Banik N, Eliceiri KW, Gopal DV. Development of a Low-Cost Gastroscope Prototype (GP) for Potential Cost-Effective Gastric Cancer Screening in Prevalent Regions. JOURNAL OF DIGESTIVE ENDOSCOPY 2023. [DOI: 10.1055/s-0043-1762574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Abstract
Background Screening for gastric cancer is known to be associated with reduced mortality in populations with high prevalence. However, many countries with high prevalence do not screen, with high costs being a significant reason for this.
Aims To describe, develop, and assess the potential for a low-cost gastroscope for early cancer screening and patient risk stratification.
Methods Our interdisciplinary team used both off-the-shelf and fabricated components to create multiple gastroscope prototypes (GP) in iterative fashion based off clinician feedback. Clinician endoscopists were surveyed using Likert scales regarding device potential, video quality, and handling when testing on a GI training device. Video quality comparison to clinically standard high-definition white light endoscopy (HD-WLE) was done using the absolute categorical ratings (ACR) method.
Results A candidate cost-effective GP with clinical potential was developed. Although initial versions were scored as inferior via ACR on all views tested when compared to HD-WLE (p < 0.001), participants agreed the concept may be beneficial (M = 4.52/5, SD = 0.72). In testing improved versions, participants agreed the device had the ability to identify discrete (M = 4.62/5, SD = 0.51) and subtle lesions (M = 4/5, SD = 0.7) but most felt video quality, although improved, was still less than HD-WLE. Sufficiency of maneuverability of device to visualize gastric views was rated as equivocal (M = 2.69/5, SD = 1.25). Conclusion The presented low-cost gastroscopic devices have potential for clinical application. With further device development and refinement including the possible addition of technologies in telemedicine and artificial intelligence, we hope the GP can help expand gastric cancer screening for populations in need.
Collapse
Affiliation(s)
- James Xu
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
- Department of Internal Medicine, Kaiser Permanente San Francisco Medical Center, University of California-San Francisco, and University of California-Berkeley, United States
| | - Mark E. Benson
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Liam M. Granlund
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Seth Gehrke
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Dylan Stanfield
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Jennifer Weiss
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Patrick Pfau
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Anurag Soni
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Ben L. Cox
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - George Petry
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Robert A. Swader
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Mark Reichelderfer
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Zhanhai Li
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Wisconsin, United States
| | - Tenzin Atrukstang
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Nyah Banik
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| | - Kevin W. Eliceiri
- Morgridge Institute for Research, Madison, Wisconsin, United States
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Wisconsin, United States
| | - Deepak V. Gopal
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Wisconsin, United States
| |
Collapse
|
5
|
Wong MCS, Deng Y, Huang J, Bai Y, Wang HHX, Yuan J, Zhang L, Yip HC, Chiu PWY. Performance of screening tests for esophageal squamous cell carcinoma: a systematic review and meta-analysis. Gastrointest Endosc 2022; 96:197-207.e34. [PMID: 35413332 DOI: 10.1016/j.gie.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/04/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS This systematic review and meta-analysis aims to compare the pooled diagnostic accuracy of the currently available esophageal squamous cell carcinoma (ESCC) screening tests. METHODS A comprehensive literature search of Embase and Medline (up to October 31, 2020) was performed to identify eligible studies. We pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio for ESCC screening tools using a bivariate random-effects model. The summary receiver operating characteristic curves with area under the curve (AUC) were plotted for each screening test. RESULTS We included 161 studies conducted in 81 research articles involving 32,209 subjects. The pooled sensitivity, specificity, and AUC of the major screening tools were respectively as follows: endoscopy (peroral endoscopy): .94 (95% confidence interval [CI], .87-.97), .92 (95% CI, .87-.95), and .97 (95% CI, .96-.99); endoscopy (transnasal endoscopy): .85 (95% CI, .70-.93), .96 (95% CI, .91-.98), and .97 (95% CI, .95-.98); microRNA: .77 (95% CI, .75-.80), .78 (95% CI, .75-.80), and .85 (95% CI, .81-.87); autoantibody: .45 (95% CI, .36-.53), .91 (95% CI, .89-.93), and .84 (95% CI, .81-.87); and cytology: .82 (95% CI, .60-.93), .97 (95% CI, .88-.99), and .97 (95% CI, .95-.98). There was high heterogeneity. CONCLUSIONS The diagnostic accuracy seemed to be comparable between cytology and endoscopy, whereas autoantibody and microRNAs bear potential as future noninvasive screening tools for ESCC. To reduce ESCC-related death in high-risk populations, it is important to develop a more accurate and less-invasive screening test.
Collapse
Affiliation(s)
- Martin C S Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China; School of Public Health, Peking Union Medical College and The Chinese Academy of Medical Sciences, Beijing, China; Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Yunyang Deng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yijun Bai
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry H X Wang
- School of Public Health, Sun Yat-Sen University, Guangzhou, China; General Practice and Primary Care, Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jinqiu Yuan
- Clinical Research Centre, Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Lin Zhang
- School of Public Health, Peking Union Medical College and The Chinese Academy of Medical Sciences, Beijing, China; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hon Chi Yip
- Department of Surgery, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Philip Wai Yan Chiu
- Department of Surgery, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Waters JK, Reznik SI. Update on Management of Squamous Cell Esophageal Cancer. Curr Oncol Rep 2022; 24:375-385. [PMID: 35142974 DOI: 10.1007/s11912-021-01153-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/26/2022]
Abstract
PURPOSE OF THE REVIEW Esophageal cancer is the sixth most common cause of cancer death globally. Squamous cell carcinoma of the esophagus (ESCC) is the predominant histologic type in the world. Treatment strategies have evolved in the last decade and new paradigms are replacing traditional approaches at all stages of cancer. This review will summarize the epidemiology, diagnosis, staging, and treatment of esophageal squamous cell carcinoma. RECENT FINDINGS Novel approaches to screening may be cost-effective in regions with a high incidence of ESCC. Multi-disciplinary evaluation and treatment has become the standard of care. Endoscopic resection may be an option for early stage ESCC. Minimally invasive esophagectomy can be performed safely as a primary therapy or after-induction chemoradiation. Several recent studies have found a survival benefit to immunotherapy for patients with metastatic or persistent disease. Multi-disciplinary evaluation and multi-modal therapy including cytotoxic chemotherapy, radiation, surgery, and immunotherapy have improved survival compared to surgery alone.
Collapse
Affiliation(s)
- John K Waters
- Division of Thoracic Surgery, Department of Cardiovascular and Thoracic Surgery, 5323 Harry Hines Boulevard, MC 8879, Dallas, TX, 75390-8879, USA
| | - Scott I Reznik
- Division of Thoracic Surgery, Department of Cardiovascular and Thoracic Surgery, 5323 Harry Hines Boulevard, MC 8879, Dallas, TX, 75390-8879, USA.
| |
Collapse
|
7
|
RISSO MFA, COSTA LCDS, TERCIOTI JR V, FERRER JAP, LOPES LR, ANDREOLLO NA. THE ESOPHAGEAL, GASTRIC, AND COLORECTAL TUMORS AND THE ESOPHAGOGASTRODUODENOSCOPIES AND COLONOSCOPIES BY THE BRAZILIAN UNIFIED HEALTH SYSTEM: WHAT IS THE IMPORTANCE? ABCD. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA (SÃO PAULO) 2022; 35:e1661. [PMID: 35766606 PMCID: PMC9254608 DOI: 10.1590/0102-672020210002e1661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/06/2021] [Indexed: 12/24/2022]
Abstract
Esophagogastroduodenoscopies and colonoscopies are the main diagnostic
examinations for esophageal, stomach, and colorectal tumors.
Collapse
|
8
|
Histórico familiar de câncer gástrico em pacientes dispépticos indicados à triagem endoscópica. ACTA PAUL ENFERM 2021. [DOI: 10.37689/acta-ape/2021ao001985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
9
|
Hu R, Bi R, Jiang L, Yang X, Zhong Y, Xie X. LncRNA TUSC8 suppresses the proliferation and migration of esophageal cancer cells by downregulation of VEGFA. J Cancer 2021; 12:6393-6400. [PMID: 34659529 PMCID: PMC8489135 DOI: 10.7150/jca.57814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/21/2021] [Indexed: 02/07/2023] Open
Abstract
Objective: This study aims to determine the expression pattern of long non-coding RNA (lncRNA) TUSC8 in esophageal cancer tissues and cell lines, to investigate its effects on esophageal cancer cell proliferation and migration, and to explore the mechanism of TUSC8-mediated esophageal cancer suppression via VEGFA downregulation. Patients and Methods: TUSC8 levels in esophageal cancer tissues and cell lines were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The influence of TUSC8 on clinical features in esophageal cancer patients was analyzed. After intervening TUSC8 expression in esophageal cancer cells, the proliferative and migratory abilities were examined in OE19 and TE-1 cells through a series of function experiments. The interaction between TUSC8 and VEGFA was assessed by the bioinformatics prediction and dual-luciferase reporter assay. Finally, the co-regulation of TUSC8 and VEGFA on esophageal cancer cell functions was evaluated. Results: TUSC8 was downregulated in esophageal cancer tissues compared with normal ones. Identically, decreased TUSC8 expression was detected in esophageal cancer cell lines compared with control cells. Low TUSC8 expression predicted poor prognosis in patients with esophageal cancer. Knockdown of TUSC8 promoted the proliferative and migratory abilities in OE19 cells, whereas overexpression of TUSC8 resulted in opposite results in TE-1 cells. VEGFA was confirmed to be a target gene of TUSC8. Overexpression of VEGFA could reverse the regulatory effects of TUSC8 on esophageal cancer cell proliferation and migration. Conclusions: LncRNA TUSC8 is downregulated in esophageal cancer tissues and cell lines. TUSC8 inhibits the proliferative and migratory abilities in esophageal cancer cells in vitro by negatively regulating VEGFA.
Collapse
Affiliation(s)
- Rui Hu
- Department of Cardiothoracic Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Rui Bi
- Department of Cardiothoracic Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Lianyong Jiang
- Department of Cardiothoracic Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xuhui Yang
- Department of Cardiothoracic Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuan Zhong
- Department of Cardiothoracic Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiao Xie
- Department of Cardiothoracic Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
10
|
Benbassat J. Sharing With Patients the Uncertainties Regarding the Management of Dyspepsia. Front Med (Lausanne) 2021; 8:681587. [PMID: 34604249 PMCID: PMC8481578 DOI: 10.3389/fmed.2021.681587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/25/2021] [Indexed: 11/25/2022] Open
Abstract
Background: The management of patients with dyspepsia is uncertain. Some authors advocate endoscopy for all; others restrict endoscopy only to patients at high risk of gastric cancer, namely to those above an age threshold, or with a family history, dysphagia, loss of weight, anemia, or a childhood in Asian countries. Still others recommend various combinations between test-and-treat for Helicobacter pylori, anti-secretory treatment, and/or endoscopy. Objective: To highlight the uncertainties in the choice between the various strategies and argue that these uncertainties should be shared with the patient. Method: An overview of reported life expectancy, patient satisfaction, gastric cancer detection rates, symptom relief, and cost effectiveness of the management strategies for dyspepsia. Main Findings: There are no randomized controlled trials of the effect of screening by endoscopy on mortality of patients with gastric cancer. Lower grades of evidence suggest that early diagnosis reduces this mortality. Analyses, which assume a survival benefit of early diagnosis, indicate that mass screening in countries of high incidence gastric cancer (> 10 cases per 100,000) and targeted screening of high-risk persons in countries of low-intermediate incidence (<10 cases per 100,000) is cost-effective at a willingness to pay of $20,000–50,000 per QALY. Prompt endoscopy appears to be best for patient satisfaction and gastric cancer detection, and test-and-treat for H pylori—for symptom relief and avoiding endoscopies. Conclusions: The gain in life expectancy is the main source of uncertainty in the choice between management strategies. This choice should be shared with the patients after explaining uncertainties and eliciting their preferences.
Collapse
Affiliation(s)
- Jochanan Benbassat
- Department of Medicine, Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
11
|
Habbous S, Yermakhanova O, Forster K, Holloway CMB, Darling G. Variation in Diagnosis, Treatment, and Outcome of Esophageal Cancer in a Regionalized Care System in Ontario, Canada. JAMA Netw Open 2021; 4:e2126090. [PMID: 34546371 PMCID: PMC8456383 DOI: 10.1001/jamanetworkopen.2021.26090] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IMPORTANCE Esophageal cancer remains one of the most deadly cancers, ranking sixth highest among cancers leading to the greatest years of life lost. OBJECTIVE To determine how patients with esophageal cancer are diagnosed and treated in Ontario's regionalized thoracic surgery centers. DESIGN, SETTING, AND PARTICIPANTS This cohort study included patients diagnosed with esophageal cancer between January 1, 2010, and December 31, 2018, identified from the Ontario Cancer Registry, in a single-payer health care system with regionalization of thoracic surgery in the province of Ontario, Canada. EXPOSURES Exposures included incidence of esophageal cancer and stage at diagnosis; time from the first health care visit until treatment; and the use of specialist consultations, endoscopic ultrasonography, positron emission tomography and computed tomography, endomucosal resection, esophagectomy, neoadjuvant therapy, adjuvant therapy, radiation alone, and chemotherapy alone or in combination with other treatment. MAIN OUTCOMES AND MEASURES Outcome measures included wait times, health care use, treatment, and overall survival. Data were analyzed from March 2020 to February 2021. RESULTS There were 10 364 patients (mean [SD] age, 68.3 [11.9] years; 7876 men [76%]) identified during the study period. The incidence of esophageal cancer increased over the study period from 1041 in 2010 to 1309 in 2018, which was driven by a 30% increase in the number of adenocarcinomas. The time from first health care encounter to start of treatment was a median 93 days (interquartile range, 56-159 days). Endoscopic ultrasonography was observed for 12% of patients, and positron emission tomography and computed tomography (CT) in 45%. Use of endoscopic mucosal resection was observed for 8% of patients with stage 0 to I disease. A total of 114 of 547 patients (21%) receiving endoscopic resection had a subsequent esophagectomy. Only 2778 patients (27%) had consultations with a thoracic surgeon, a medical oncologist, and a radiation oncologist, whereas 1514 patients (15%) did not see any of these specialists. Of 3047 patients who had an esophagectomy, those receiving neoadjuvant therapy had better overall survival (median survival, 36 months; 95% CI, 32-39 months) than patients who received esophagectomy alone (median survival, 27 months; 95% CI, 24-30 months) or those who received esophagectomy with adjuvant therapy (median survival, 36 months; 95% CI, 32-44 months) despite significant early mortality (log-rank P < .001). There was significant variation in treatment modality across hospitals: esophagectomy ranged from 5% to 39%; esophagectomy after neoadjuvant therapy ranged from 33% to 93%; and esophagectomy followed by adjuvant therapy ranged from 0 to 34% (P < .001). Perioperative mortality was higher at 30 days for patients receiving esophagectomy at low-volume centers (odds ratio [OR], 3.66; 95% CI, 2.01-6.66) and medium-volume centers (OR, 2.07; 95% CI, 1.33-3.23) compared with high-volume centers (P < .001). A longer wait time until treatment was associated with better overall survival (median overall survival was 15 to 17 days vs 5 to 8 days for patients who received treatment earlier than 30 days vs 30 days or longer after diagnosis; P < .001). CONCLUSIONS AND RELEVANCE The results of this cohort study suggest that despite regionalization, there was significant regional variability in volumes at designated centers and in the evaluation and treatment course for patients with esophageal cancer across Ontario.
Collapse
Affiliation(s)
| | | | | | - Claire M. B. Holloway
- Ontario Health (Cancer Care Ontario), Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Gail Darling
- Ontario Health (Cancer Care Ontario), Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Toronto General Hospital, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Lv Y, Lu J, Liu X, Miao S, Mao X, Li B, Pei R, Xiang C. Histone deacetylase 1 regulates the malignancy of oral cancer cells via miR-154-5p/PCNA axis. Biol Chem 2021; 401:1273-1281. [PMID: 32549181 DOI: 10.1515/hsz-2020-0189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Abstract
Histone deacetylases (HDACs) can regulate the progression of various cancers, while their roles in oral cancer cells are not well known. Our present study found that the HDAC1 was over expressed in oral squamous cell carcinoma (OSCC) cells and tissues. Targeted inhibition of HDAC1 via its specific inhibitor PCI24781 or siRNA can inhibit the proliferation of OSCC cells and increase their sensitivity to the chemo-sensitivity such as doxorubicin treatment. HDAC1 can regulate the expression of proliferating cell nuclear antigen (PCNA) via decreasing its mRNA stability. While over expression of PCNA can attenuate HDAC1 inhibition induced suppression of cell proliferation. We checked the expression of various miRNAs which can target the 3'UTR of PCNA. Results showed that HDAC1 can negative regulate the expression of miR-154-5p, inhibitor of miR-154-5p can attenuate PCI24781 treatment decreased PCNA expression and cell proliferation. Collectively, our present study suggested that HDAC1 can promote the growth and progression of OSCC via regulation of miR-154-5p/PCNA signals.
Collapse
Affiliation(s)
- Yuanjing Lv
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Jinle Lu
- Department of Head and Neck Thyroid Surgery, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Hebei, Cangzhou 061000, China
| | - Xin Liu
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Xionghui Mao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Baojun Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Rong Pei
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Cheng Xiang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, China
| |
Collapse
|
13
|
Planade O, Dessomme B, Chapelle N, Verdier M, Duchalais E, Queneherve L, Le Rhun M, Coron E, Mosnier JF, Matysiak-Budnik T, Touchefeu Y. Systematic upper endoscopy concomitant with colonoscopy performed within the colorectal cancer screening program: Impact on the patients' management. Clin Res Hepatol Gastroenterol 2021; 45:101501. [PMID: 33714864 DOI: 10.1016/j.clinre.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS The French colorectal cancer screening program is based on a fecal immunochemical test, followed by colonoscopy in case of positivity. The benefit of adding a concomitant upper endoscopy to detect upper digestive lesions (precancerous or others) is still debated. Our aim was to evaluate the frequency of upper digestive lesions detected by upper endoscopy performed concomitantly with colonoscopy following a positive fecal immunochemical test, and their impact on the patients' management (i.e., surveillance, medical treatment, endoscopic or surgical procedure). METHODS Data of all the patients who consulted for a positive test between May 2016 and May 2019 in our center, and for whom concomitant upper endoscopy and colonoscopy were performed, were analyzed retrospectively. Patients with significant history of upper gastrointestinal diseases or with current gastrointestinal symptoms were excluded. RESULTS One hundred patients were included [median age (min-max): 62 (50-75), men 64%]. Macroscopic and/or microscopic upper digestive lesions were found in 58 of them (58%): Helicobacter pylori infection in 17 patients, gastric precancerous lesions in 9 patients (chronic atrophic gastritis with intestinal metaplasia, n=8, low grade dysplasia, n=1), Barrett's esophagus requiring surveillance in 4 patients, and 1 duodenal adenoma with low-grade dysplasia. In 44 patients (44%), the upper endoscopy findings had an impact on patients' management, with no significant difference between the groups with positive (CRC or advanced adenoma)- or negative (any other lesions or normal) colonoscopy. CONCLUSION A systematic upper endoscopy combined with colonoscopy for positive fecal immunochemical test could represent an efficient strategy for upper digestive lesions screening in France. Further studies are necessary to confirm these results and to evaluate cost-effectiveness of this approach.
Collapse
Affiliation(s)
- Orianne Planade
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Brigitte Dessomme
- Service d'Evaluation Médicale et d'Epidémiologie, Nantes University Hospital, 85 rue Saint-Jacques, Nantes 44093, France
| | - Nicolas Chapelle
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Marine Verdier
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Emilie Duchalais
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Lucille Queneherve
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Marc Le Rhun
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Emmanuel Coron
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Jean-Francois Mosnier
- Service d'anatomie et cytologie pathologiques, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| | - Tamara Matysiak-Budnik
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France.
| | - Yann Touchefeu
- Institut des Maladies de l'appareil Digestif, Hépato-Gastroentérologie & Oncologie Digestive, Hôtel Dieu, Nantes University Hospital, 1 place Alexis Ricordeau, Nantes 44093, France
| |
Collapse
|
14
|
Khaitan PG, El-Sayed Abbas A, Watson TJ. Underfunding of Esophageal Cancer Research. Ann Thorac Surg 2020; 111:1099-1101. [PMID: 33359725 DOI: 10.1016/j.athoracsur.2020.09.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/19/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Puja Gaur Khaitan
- Department of Surgery, Division of Thoracic and Esophageal Surgery, Georgetown University School of Medicine, MedStar Washington Hospital Center, Washington, DC.
| | - Abbas El-Sayed Abbas
- Department of Thoracic Medicine and Surgery, Division of Thoracic Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Thomas J Watson
- Department of Surgery, Division of Thoracic and Esophageal Surgery, Georgetown University School of Medicine, MedStar Washington Hospital Center, Washington, DC
| |
Collapse
|
15
|
Lee NR, Meng RY, Rah SY, Jin H, Ray N, Kim SH, Park BH, Kim SM. Reactive Oxygen Species-Mediated Autophagy by Ursolic Acid Inhibits Growth and Metastasis of Esophageal Cancer Cells. Int J Mol Sci 2020; 21:E9409. [PMID: 33321911 PMCID: PMC7764507 DOI: 10.3390/ijms21249409] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Ursolic acid (UA) possesses various pharmacological activities, such as antitumorigenic and anti-inflammatory effects. In the present study, we investigated the mechanisms underlying the effects of UA against esophageal squamous cell carcinoma (ESCC) (TE-8 cells and TE-12 cells). The cell viability assay showed that UA decreased the viability of ESCC in a dose-dependent manner. In the soft agar colony formation assay, the colony numbers and size were reduced in a dose-dependent manner after UA treatment. UA caused the accumulation of vacuoles and LC3 puncta, a marker of autophagosome, in a dose-dependent manner. Autophagy induction was confirmed by measuring the expression levels of LC3 and p62 protein in ESCC cells. UA increased LC3-II protein levels and decreased p62 levels in ESCC cells. When autophagy was hampered using 3-methyladenine (3-MA), the effect of UA on cell viability was reversed. UA also significantly inhibited protein kinase B (Akt) activation and increased p-Akt expression in a dose-dependent manner in ESCC cells. Accumulated LC3 puncta by UA was reversed after wortmannin treatment. LC3-II protein levels were also decreased after treatment with Akt inhibitor and wortmannin. Moreover, UA treatment increased cellular reactive oxygen species (ROS) levels in ESCC in a time- and dose-dependent manner. Diphenyleneiodonium (an ROS production inhibitor) blocked the ROS and UA induced accumulation of LC3-II levels in ESCC cells, suggesting that UA-induced cell death and autophagy are mediated by ROS. Therefore, our data indicate that UA inhibits the growth of ESCC cells by inducing ROS-dependent autophagy.
Collapse
Affiliation(s)
- Na-Ri Lee
- Division of Hematology and Oncology, Jeonbuk National University Medical School, Jeonju 54907, Korea;
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea;
- Research Institute of Clinical Medicine, Biomedical Research Institute of Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Ruo Yu Meng
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| | - So-Young Rah
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea; (S.-Y.R.); (B.H.P.)
| | - Hua Jin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China;
| | - Navin Ray
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| | - Seong-Hun Kim
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| | - Byung Hyun Park
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea; (S.-Y.R.); (B.H.P.)
| | - Soo Mi Kim
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| |
Collapse
|
16
|
Wu D, Li H, Wang J, Li H, Xiao Q, Zhao X, Huo Z. LncRNA NEAT1 promotes gastric cancer progression via miR-1294/AKT1 axis. Open Med (Wars) 2020; 15:1028-1038. [PMID: 33336058 PMCID: PMC7718639 DOI: 10.1515/med-2020-0218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/17/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) were reported to promote the development of gastric cancer (GC). Nuclear-enriched abundant transcript 1 (NEAT1) played a great role in diverse cancers, but the mechanism of NEAT1 in GC remains indistinct. NEAT1 and AKT1 were distinctly up-regulated and miR-1294 was down-regulated in GC tissues and cells. Cell proliferation and metastasis were refrained but apoptosis was promoted in GC cells after knockdown of NEAT1. NEAT1 negatively regulated miR-1294 expression, and the miR-1294 inhibitor reverted the si-NEAT1-induced effect on GC cells. NEAT1 modulated AKT1 expression through miR-1294, and the si-NEAT1-induced effect was relieved by AKT1. NEAT1 affected phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway via regulating miR-1294 and AKT1. NEAT1 could modulate cell proliferation, apoptosis, and metastasis in GC cells by regulating the PI3K/AKT/mTOR signaling pathway via the miR-1294/AKT1 axis, showing the great potential for NEAT1 as a valid biomarker in the progression and treatment of GC.
Collapse
Affiliation(s)
- Dianchao Wu
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Hui Li
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Junfeng Wang
- Department of Colorectal Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Li
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Qihai Xiao
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Xiaofeng Zhao
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Zhibin Huo
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| |
Collapse
|
17
|
Liu L, Liu QW, Wu XD, Liu SY, Cao HJ, Hong YT, Qin HY. Follow-up study on symptom distress in esophageal cancer patients undergoing repeated dilation. World J Clin Cases 2020; 8:3503-3514. [PMID: 32913857 PMCID: PMC7457119 DOI: 10.12998/wjcc.v8.i16.3503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/03/2020] [Accepted: 07/14/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Repeated endoscopic probe dilatation is the most preferred treatment for esophageal stenosis which may cause high levels of symptom distress in the patient's home rehabilitation stage.
AIM To explore the changes in the symptom distress level and its correlation with the dilation effect in patients with esophageal carcinoma undergoing repeated dilations for lumen stenosis.
METHODS The difference (R2-R1) between the diameter of the esophageal stenosis opening (R1) of the patients before dilation (R1) and after dilation (R2) was calculated to describe the extent and expansion of the esophageal stenosis before and after dilation. The M.D. Anderson Symptom Inventory was used to describe the symptom distress level of patients with dilation intermittence during their stay at home and to explore the correlation between the dilation effect and symptom distress level.
RESULTS The diameter of the esophagus (R1) increased before each dilation in patients undergoing esophageal dilation (P < 0.05). The diameter (R2) increased after dilation (P < 0.05); the dilation effect (R2-R1) decreased with the number of dilations (P < 0.05). The total symptom distress score significantly increased with the number of dilations (P < 0.05). The symptom distress scores of the patients were negatively correlated (P < 0.05) with the previous dilation effect (R2-R1) and the esophageal diameter (R2) after the previous dilation. After the 1st to 4th dilations, the patient's symptom distress score was negatively correlated with the esophageal diameter (R12) before the next dilation, while there was no significant correlation (P > 0.05) with the other dilations.
CONCLUSION In patients who have undergone repeated dilations, better effect stands for lower symptom distress level and the increase in symptom distress has a prompt effect on the severity of the next occurrence of restenosis.
Collapse
Affiliation(s)
- Li Liu
- Thoracic Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Qian-Wen Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xiao-Dan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Shu-Yue Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Hepatopancreatobiliary Surgery Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hui-Jiao Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Traditional Chinese Medicine Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Yu-Tong Hong
- Thoracic Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Hui-Ying Qin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Nursing, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
18
|
Yao W, Guo P, Mu Q, Wang Y. Exosome-Derived Circ-PVT1 Contributes to Cisplatin Resistance by Regulating Autophagy, Invasion, and Apoptosis Via miR-30a-5p/YAP1 Axis in Gastric Cancer Cells. Cancer Biother Radiopharm 2020; 36:347-359. [PMID: 32799541 DOI: 10.1089/cbr.2020.3578] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: Emerging studies manifested that exosomal RNAs had pivotal roles in human cancer therapies. This article aimed to research the regulatory mechanism of exosomal circRNA-plasmacytoma variant translocation 1 (circ-PVT1) in cisplatin (DDP) resistance of gastric cancer (GC). Methods: Exosomes were isolated by ExoQuick® method and ultracentrifugation and then identified through transmission electron microscope and the examination of exosome markers. Related proteins were detected using Western blot. Quantitative real-time polymerase chain reaction (qRT-PCR) was applied for measuring circ-PVT1, microRNA-30a-5p (miR-30a-5p), and Yes-associated protein 1 (YAP1) expression. The half inhibitory concentration (IC50) of DDP was assessed by 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide (MTT). Cell apoptosis and invasion were, respectively, determined using flow cytometry and transwell assay. Target relationship was confirmed by dual-luciferase reporter assay. The impact of circ-PVT1 on DDP resistance was explored via xenograft tumor assay. Results: Exosomal circ-PVT1 was upregulated while miR-30a-5p was downregulated in DDP-resistant GC serums and cells. Circ-PVT1 knockdown repressed DDP resistance in DDP-resistant GC cells via promoting apoptosis and decreasing invasion or autophagy by negatively targeting miR-30a-5p. YAP1 was a direct target of miR-30a-5p. MiR-30a-5p overexpression inhibited DDP resistance via reducing YAP1. Circ-PVT1 modulated YAP1 expression by targeting miR-30a-5p. Circ-PVT1 depression expedited DDP sensitivity of GC via miR-30a-5p/YAP1 axis in vivo. Conclusion: Exosomal circ-PVT1 facilitated DDP resistance via modulating autophagy, invasion and apoptosis by miR-30a-5p/YAP1 axis in GC cells. Exosomal circ-PVT1 might be a prospective indicator in DDP therapy of GC.
Collapse
Affiliation(s)
- Wenjuan Yao
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Peng Guo
- Department of Hepatology and Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Qiming Mu
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuecheng Wang
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
19
|
Experimental Study of the Effect of Helicobacter pylori Infection on Barrett Esophagus and Its Correlation with Immune Function. Jundishapur J Microbiol 2020. [DOI: 10.5812/jjm.98422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Helicobacter pylori is an important pathogen in the upper digestive tract. It is of great significance to properly understand the risk factors for the transformation of Barrett esophagus into esophageal carcinoma. However, the relationship between H. pylori and gastroesophageal reflux disease (GERD) and Barrett esophagus remains controversial, and the correlation with immune function has been rarely reported. Objectives: This study investigated the effect of H. pylori infection on Barrett esophagus and its correlation with immune function. Methods: We recruited 40 patients with Barrett esophagus (Barrett esophagus group) and 40 patients with GERD (GERD group). In addition, 40 healthy controls were selected for the control group. Esophageal function and its correlation with immune function were measured in each group. Results: The positivity rate of H. pylori (P < 0.05) and sphincter pressure were lower in both Barrett esophagus and GERD groups than in the control group, while the levels of PGI, PGII, PGI/II, and G-17 were higher (P < 0.05). The levels of CD3+, CD4+, and CD4+/CD8+ were lower in the Barrett esophagus group than in the GERD group, but they were negatively correlated (P < 0.05) with H. pylori infection. The level of CD8+ was higher in the Barrett esophagus group, and it was positively correlated (P < 0.05) with H. pylori infection. Conclusions: Helicobacter pylori infection may protect against Barrett esophagus by reducing gastric acid secretion and increasing lower esophageal sphincter pressure. Besides, it has a certain correlation with immune function.
Collapse
|
20
|
Zhang G, Liu Y, Dong F, Liu X. Transcription/Expression of KLRB1 Gene as A Prognostic Indicator in Human Esophageal Squamous Cell Carcinoma. Comb Chem High Throughput Screen 2020; 23:667-674. [PMID: 32416673 DOI: 10.2174/1386207323666200517114154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/07/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022]
Abstract
AIM AND OBJECTIVE Esophageal squamous cell carcinoma (ESCC) is the most prevalent type of cancer with worldwide distribution and dismal prognosis despite ongoing efforts to improve treatment options. Therefore, it is essential to determine the prognostic factors for ESCC. METHODS AND RESULTS We determined KLRB1 to be a prognostic indicator of human ESCC. KLRB1 was expressed at low levels in ESCC patients. Based on the risk score, patients were divided into high and low-risk groups. High-risk patients showed a poor survival rate. The prediction model based on the N stage, sex, and KLRB1 was significantly better than that based on the N stage and sex. The modified prediction model showed a robust ROC curve with an AUC value of 0.973. The knockdown of KLRB1 inhibited the growth of human ESCC cells. KLRB1 regulated Akt, mTOR, p27, p38, NF-κB, Cyclin D1, and JNK signaling, which was consistent with the result of GSEA. CONCLUSION KLRB1 is a potential prognostic marker for human ESCC patients.
Collapse
Affiliation(s)
- Guangwei Zhang
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Ying Liu
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Fajin Dong
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Xianming Liu
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| |
Collapse
|